1
|
Bertoncello KT, Bonan CD. The Effect of Adenosine Signaling on Memory Impairment Induced by Pentylenetetrazole in Zebrafish. Neurochem Res 2023; 48:1889-1899. [PMID: 36729312 DOI: 10.1007/s11064-023-03867-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 02/03/2023]
Abstract
Epilepsy is characterized by the manifestation of spontaneous and recurrent seizures. The high prevalence of comorbidities associated with epilepsy, such as cognitive dysfunction, affects the patients quality of life. Adenosine signaling modulation might be an effective alternative to control seizures and epilepsy-associated comorbidities. This study aimed to verify the role of adenosine modulation on the seizure development and cognitive impairment induced by pentylenetetrazole (PTZ) in zebrafish. At first, animals were submitted to a training session in the inhibitory avoidance test and, after 10 min, they received an intraperitoneal injection of valproate, adenosine A1 receptor agonist cyclopentyladenosine (CPA), adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), adenosine A2A receptor antagonist ZM 241385, adenosine deaminase inhibitor erythro-9-(2-hydroxy-3-nony1)-adenine hydrochloride (EHNA) or the nucleoside transporter inhibitor dipyridamole. Thirty min after the intraperitoneal injection, the animals were exposed to 7.5 mM PTZ for 10 min, where they were evaluated for latency to reach the seizure stages (I, II, and III). Finally, 24 h after the training session, the animals were submitted to the inhibitory avoidance test to verify their cognitive performance during the test session. Valproate, CPA, and EHNA showed antiseizure effects and prevented the memory impairment induced by PTZ exposure. DPCPX, ZM 241385, and dipyridamole pretreatments caused no changes in seizure development; however, these drugs prevented memory impairment without altering locomotion. Our results reinforce the antiseizure effects of adenosine signaling and support the idea that the involvement of adenosine in memory processes may be a target for preventive strategies against cognitive impairment associated with epilepsy.
Collapse
Affiliation(s)
- Kanandra Taisa Bertoncello
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil. .,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
2
|
Tang Y, Sun L, Zhao Y, Yao J, Feng Z, Liu Z, Zhang G, Sun C. UHPLC-ESI-QE-Orbitrap-MS based metabolomics reveals the antioxidant mechanism of icaritin on mice with cerebral ischemic reperfusion. PeerJ 2023; 11:e14483. [PMID: 36643627 PMCID: PMC9838208 DOI: 10.7717/peerj.14483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/08/2022] [Indexed: 01/12/2023] Open
Abstract
Background Icaritin (ICT) has been previously demonstrated to display protective effects against cerebral ischemic reperfusion (I/R) by inhibiting oxidative stress, but the mechanism remains unclear. This study aimed to explore the mechanism from the perspective of metabolomics. Methods A mice cerebral artery occlusion/reperfusion (MCAO/R) model was explored to mimic cerebral ischemic reperfusion and protective effect of ICT was assessed by neurologic deficit scoring, infarct volume and brain water content. Ultra-high-performance liquid chromatography electrospray ionization orbitrap tandem mass spectrometry (UHPLC-ESI-QE-Orbitrap-MS) based metabolomic was performed to explore potential biomarkers. Brain tissue metabolic profiles were analyzed and metabolic biomarkers were identified through multivariate data analysis. The protein levels of Nrf2, HO-1 and HQO1 were assayed by western blot. The release of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT) were detected using corresponding assay kits. Results The results showed that after ICT treatment, the neurological deficit, cerebral infarction area, brain edema and the level of MDA in brain tissue of MCAO/R mice were significantly reduced. Meanwhile, ICT enhanced the activity of SOD, CAT and GSH-Px. Western blot results confirmed that ICT up-regulated the protein levels of antioxidant-related protein including Nrf2, HO-1 and NQO1. According to the metabolomic profiling of brain tissues, clear separations were observed among the Sham, Model and ICT groups. A total of 44 biomarkers were identified, and the identified biomarkers were mainly related to linoleic acid metabolism, arachidonic acid metabolism, alanine, aspartate and glutamate metabolism, arginine biosynthesis, arginine and proline metabolism, D-glutamine and D-glutamate metabolism, taurine and hypotaurine metabolism and purine metabolism, respectively. At the same time, the inhibitory effect of ICT on arachidonic acid and linoleic acid in brain tissue, as well as the promoting effect on taurine, GABA, NAAG, may be the key factors for the anti-neurooxidative function of mice after MCAO/R injury. Conclusion Our results demonstrate that ICT has benefits for MCAO/R injury, which are partially related to the suppression of oxidative stress via stimulating the Nrf2 signaling and regulating the production of arachidonic acid, linoleic acid, taurine, GABA, NAAG in brain tissue.
Collapse
Affiliation(s)
- Yunfeng Tang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Lixin Sun
- Linyi Traditional Chinese Medicine Hospital, Linyi, Shandong Province, China
| | - Yun Zhao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Jingchun Yao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Zhong Feng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China,School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Zhong Liu
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| | - Chenghong Sun
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, Shandong Province, China
| |
Collapse
|
3
|
Yan X, Zhao X, Zhou Z, McKay A, Brunet A, Zare RN. Cell-Type-Specific Metabolic Profiling Achieved by Combining Desorption Electrospray Ionization Mass Spectrometry Imaging and Immunofluorescence Staining. Anal Chem 2020; 92:13281-13289. [PMID: 32880432 PMCID: PMC8782277 DOI: 10.1021/acs.analchem.0c02519] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell-type-specific metabolic profiling in tissue with heterogeneous composition has been of great interest across all mass spectrometry imaging (MSI) technologies. We report here a powerful new chemical imaging capability in desorption electrospray ionization (DESI) MSI, which enables cell-type-specific and in situ metabolic profiling in complex tissue samples. We accomplish this by combining DESI-MSI with immunofluorescence staining using specific cell-type markers. We take advantage of the variable frequency of each distinct cell type in the lateral septal nucleus (LSN) region of mouse forebrain. This allows computational deconvolution of the cell-type-specific metabolic profile in neurons and astrocytes by convex optimization-a machine learning method. Based on our approach, we observed 107 metabolites that show different distributions and intensities between astrocytes and neurons. We subsequently identified 23 metabolites using high-resolution mass spectrometry (MS) and tandem MS, which include small metabolites such as adenosine and N-acetylaspartate previously associated with astrocytes and neurons, respectively, as well as accumulation of several phospholipid species in neurons which have not been studied before. Overall, this method overcomes the relatively low spatial resolution of DESI-MSI and provides a new platform for in situ metabolic investigation at the cell-type level in complex tissue samples with heterogeneous cell-type composition.
Collapse
Affiliation(s)
- Xin Yan
- Department of Chemistry, Texas A&M University, College Station, TX 77843.; Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Xiaoai Zhao
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Zhenpeng Zhou
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Andrew McKay
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Anne Brunet
- Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Richard N. Zare
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Tang J, Jiang X, Zhou Y, Dai Y. Effects of A2BR on the biological behavior of mouse renal fibroblasts during hypoxia. Mol Med Rep 2015; 11:4397-402. [PMID: 25672943 DOI: 10.3892/mmr.2015.3320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 01/21/2015] [Indexed: 11/06/2022] Open
Abstract
Fibroblasts are the effector cells of collagen secretion in renal interstitial fibrosis (RIF), and their proliferation and activation are essential for the development of RIF. Hypoxic ischemia in local tissues has been identified in chronic kidney diseases (CKDs), with adenosine (ADO) as a key signaling molecule. The current study investigated the association between ADO and the biological behavior of renal fibroblasts by establishing an in vitro hypoxia cell model. This aimed to provide experimental evidence for the prevention and treatment of RIF. NIH3T3 fibroblasts were exposed to hypoxia, and the subtypes of the ADO receptor (AR) on the cell surface were identified by a TaqMan probe‑based assay. Cells were divided into the following four groups: i) Control; ii) 5'‑N‑ethylcarboxamidoadenosine (NECA); iii) PT, NECA + 8‑phenyltheophylline (PT); and iv) MRS, NECA + N‑(4‑cyanophenyl)‑2‑[4‑(2,3,6,7‑tetrahydro‑2,6‑dioxo‑1,3‑dipropyl‑1H‑purin‑8‑yl)phenoxy]‑acetamide (MRS1754). The mRNA levels of transforming growth factor‑β1 (TGF‑β1), procollagen α1 (I) and α‑smooth muscle actin (α‑SMA) were measured following 24, 48, and 72 h of hypoxia. Cell proliferation was evaluated by a 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide assay at 0, 12, 24, 48 and 72 h. The results demonstrated that A2BR was the predominant AR subtype present in hypoxia‑stimulated fibroblasts. NECA significantly induced fibroblast proliferation and upregulated the expression of TGF‑β1, procollagen α1 (I) and α‑SMA mRNA, while 8‑PT and MRS1754 inhibited fibroblast proliferation and downregulated the expression of TGF‑β1, procollagen α1 (I) and α‑SMA mRNA. The blockage of A2BR in hypoxia significantly inhibited the proliferation and activation of fibroblasts, and reduced the production of profibrotic cytokines, thus preventing the generation and development of fibrosis.
Collapse
Affiliation(s)
- Jin Tang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Xianzhen Jiang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yihong Zhou
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yingbo Dai
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
5
|
Burnstock G, Boeynaems JM. Purinergic signalling and immune cells. Purinergic Signal 2014; 10:529-64. [PMID: 25352330 PMCID: PMC4272370 DOI: 10.1007/s11302-014-9427-2] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 11/28/2022] Open
Abstract
This review article provides a historical perspective on the role of purinergic signalling in the regulation of various subsets of immune cells from early discoveries to current understanding. It is now recognised that adenosine 5'-triphosphate (ATP) and other nucleotides are released from cells following stress or injury. They can act on virtually all subsets of immune cells through a spectrum of P2X ligand-gated ion channels and G protein-coupled P2Y receptors. Furthermore, ATP is rapidly degraded into adenosine by ectonucleotidases such as CD39 and CD73, and adenosine exerts additional regulatory effects through its own receptors. The resulting effect ranges from stimulation to tolerance depending on the amount and time courses of nucleotides released, and the balance between ATP and adenosine. This review identifies the various receptors involved in the different subsets of immune cells and their effects on the function of these cells.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| | | |
Collapse
|
6
|
Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev 2013; 66:102-92. [PMID: 24335194 DOI: 10.1124/pr.113.008029] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purinergic signaling plays important roles in control of vascular tone and remodeling. There is dual control of vascular tone by ATP released as a cotransmitter with noradrenaline from perivascular sympathetic nerves to cause vasoconstriction via P2X1 receptors, whereas ATP released from endothelial cells in response to changes in blood flow (producing shear stress) or hypoxia acts on P2X and P2Y receptors on endothelial cells to produce nitric oxide and endothelium-derived hyperpolarizing factor, which dilates vessels. ATP is also released from sensory-motor nerves during antidromic reflex activity to produce relaxation of some blood vessels. In this review, we stress the differences in neural and endothelial factors in purinergic control of different blood vessels. The long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides in promoting migration and proliferation of both vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis and vessel remodeling during restenosis after angioplasty are described. The pathophysiology of blood vessels and therapeutic potential of purinergic agents in diseases, including hypertension, atherosclerosis, ischemia, thrombosis and stroke, diabetes, and migraine, is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; and Department of Pharmacology, The University of Melbourne, Australia.
| | | |
Collapse
|
7
|
Zhang W, Zhang Y, Wang W, Dai Y, Ning C, Luo R, Sun K, Glover L, Grenz A, Sun H, Tao L, Zhang W, Colgan SP, Blackburn MR, Eltzschig HK, Kellems RE, Xia Y. Elevated ecto-5'-nucleotidase-mediated increased renal adenosine signaling via A2B adenosine receptor contributes to chronic hypertension. Circ Res 2013; 112:1466-78. [PMID: 23584256 DOI: 10.1161/circresaha.111.300166] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
RATIONALE Hypertension is the most prevalent life-threatening disease worldwide and is frequently associated with chronic kidney disease (CKD). However, the molecular basis underlying hypertensive CKD is not fully understood. OBJECTIVE We sought to identify specific factors and signaling pathways that contribute to hypertensive CKD and thereby exacerbate disease progression. METHODS AND RESULTS Using high-throughput quantitative reverse-transcription polymerase chain reaction profiling, we discovered that the expression level of 5'-ectonucleotidase (CD73), a key enzyme that produces extracellular adenosine, was significantly increased in the kidneys of angiotensin II-infused mice, an animal model of hypertensive nephropathy. Genetic and pharmacological studies in mice revealed that elevated CD73-mediated excess renal adenosine preferentially induced A2B adenosine receptor (ADORA2B) production and that enhanced kidney ADORA2B signaling contributes to angiotensin II-induced hypertension. Similarly, in humans, we found that CD73 and ADORA2B levels were significantly elevated in the kidneys of CKD patients compared with normal individuals and were further elevated in hypertensive CKD patients. These findings led us to further discover that elevated renal CD73 contributes to excess adenosine signaling via ADORA2B activation that directly stimulates endothelin-1 production in a hypoxia-inducible factor-α-dependent manner and underlies the pathogenesis of the disease. Finally, we revealed that hypoxia-inducible factor-α is an important factor responsible for angiotensin II-induced CD73 and ADORA2B expression at the transcriptional level. CONCLUSIONS Overall, our studies reveal that angiotensin II-induced renal CD73 promotes the production of renal adenosine that is a prominent driver of hypertensive CKD by enhanced ADORA2B signaling-mediated endothelin-1 induction in a hypoxia-inducible factor-α-dependent manner. The inhibition of excess adenosine-mediated ADORA2B signaling represents a novel therapeutic target for the disease.
Collapse
Affiliation(s)
- Weiru Zhang
- Departments of Biochemistry and Molecular Biology,University of Texas Medical School at Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Komaki S, Ishikawa K, Arakawa Y. Trk and cAMP-dependent survival activity of adenosine A(2A) agonist CGS21680 on rat motoneurons in culture. Neurosci Lett 2012; 522:21-4. [PMID: 22691937 DOI: 10.1016/j.neulet.2012.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 05/27/2012] [Accepted: 06/01/2012] [Indexed: 01/01/2023]
Abstract
The survival activity of adenosine A(2A) agonist CGS21680 on motoneurons in culture through the transactivation of neurotrophin receptor TrkB has been reported previously; however, since adenosine A(2A) receptor belongs to a Gs-protein-coupled receptor, we investigated the involvement of the cAMP pathway in the survival activity of CGS21680 using purified motoneurons in culture. CGS21680 alone showed only small survival activity, but the activity was significantly enhanced by the addition of a phosphodiesterase inhibitor, IBMX. This survival activity was partially inhibited by a protein kinase A inhibitor H89 or a neurotrophin receptor tyrosine kinase inhibitor K252a, and was completely inhibited by their combination. These results indicate that the survival activity of CGS21680 on motoneurons is exerted by the mixed effect of the adenylate cyclase-cAMP-PKA pathway and transactivation of Trk neurotrophin receptor. Under conditions in which the maximum survival of motoneurons was supported by sufficient concentrations of brain-derived neurotrophic factor (BDNF), a TrkB ligand, the addition of 100μM AMPA for 3 days led to significant cell death. Treatment with CGS21680 and IBMX protected motoneurons from the toxicity of AMPA, further supporting the presence of a TrkB-independent pathway of CGS21680 activity and suggesting a novel therapeutic approach to motoneuron diseases such as amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Satoshi Komaki
- Clinical Research Support Center, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo 113-8655, Japan.
| | | | | |
Collapse
|
9
|
Measurement of purine release with microelectrode biosensors. Purinergic Signal 2011; 8:27-40. [PMID: 22095158 DOI: 10.1007/s11302-011-9273-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/19/2011] [Indexed: 12/22/2022] Open
Abstract
Purinergic signalling departs from traditional paradigms of neurotransmission in the variety of release mechanisms and routes of production of extracellular ATP and adenosine. Direct real-time measurements of these purinergic agents have been of great value in understanding the functional roles of this signalling system in a number of diverse contexts. Here, we review the methods for measuring purine release, introduce the concept of microelectrode biosensors for ATP and adenosine and explain how these have been used to provide new mechanistic insight in respiratory chemoreception, synaptic physiology, eye development and purine salvage. We finish by considering the association of purine release with pathological conditions and examine the possibilities that biosensors for purines may one day be a standard part of the clinical diagnostic tool chest.
Collapse
|
10
|
Dai Y, Zhang W, Wen J, Zhang Y, Kellems RE, Xia Y. A2B adenosine receptor-mediated induction of IL-6 promotes CKD. J Am Soc Nephrol 2011; 22:890-901. [PMID: 21511827 DOI: 10.1681/asn.2010080890] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic elevation of adenosine, which occurs in the setting of repeated or prolonged tissue injury, can exacerbate cellular dysfunction, suggesting that it may contribute to the pathogenesis of CKD. Here, mice with chronically elevated levels of adenosine, resulting from a deficiency in adenosine deaminase (ADA), developed renal dysfunction and fibrosis. Both the administration of polyethylene glycol-modified ADA to reduce adenosine levels and the inhibition of the A(2B) adenosine receptor (A(2B)R) attenuated renal fibrosis and dysfunction. Furthermore, activation of A(2B)R promoted renal fibrosis in both mice infused with angiotensin II (Ang II) and mice subjected to unilateral ureteral obstruction (UUO). These three mouse models shared a similar profile of profibrotic gene expression in kidney tissue, suggesting that they share similar signaling pathways that lead to renal fibrosis. Finally, both genetic and pharmacologic approaches showed that the inflammatory cytokine IL-6 mediates adenosine-induced renal fibrosis downstream of A(2B)R. Taken together, these data suggest that A(2B)R-mediated induction of IL-6 contributes to renal fibrogenesis and shows potential therapeutic targets for CKD.
Collapse
Affiliation(s)
- Yingbo Dai
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
11
|
Li H, Yan Z, Zhu J, Yang J, He J. Neuroprotective effects of resveratrol on ischemic injury mediated by improving brain energy metabolism and alleviating oxidative stress in rats. Neuropharmacology 2011; 60:252-8. [DOI: 10.1016/j.neuropharm.2010.09.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 08/03/2010] [Accepted: 09/06/2010] [Indexed: 01/01/2023]
|
12
|
Pajski ML, Venton BJ. Adenosine Release Evoked by Short Electrical Stimulations in Striatal Brain Slices is Primarily Activity Dependent. ACS Chem Neurosci 2010; 1:775-787. [PMID: 21218131 DOI: 10.1021/cn100037d] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adenosine is an important neuromodulator in the brain. Traditionally, adenosine is thought to arise in the extracellular space by either an extracellular mechanism, where it is formed outside the cell by the breakdown of released ATP, or an intracellular mechanism, where adenosine made inside the cell is transported out. Recently, a proposed third mechanism of activity dependent adenosine release has also been proposed. Here, we used fast-scan cyclic voltammetry to compare the time course and mechanism of adenosine formation evoked by either low- or high-frequency stimulations in striatal rat brain slices. Low-frequency stimulations (5 pulses at 10 Hz) resulted in an average adenosine efflux of 0.22 ± 0.02 μM, while high-frequency stimulations (5 pulses, 60 Hz) evoked 0.36 ± 0.04 μM. Blocking intracellular formation by inhibiting adenosine transporters with S-(4-nitrobenzyl)-6-thioinosine (NBTI) or propentofylline did not decrease release for either frequency, indicating that the release was not due to the intracellular mechanism. Blocking extracellular formation with ARL-67156 reduced low-frequency release about 60%, but did not affect high-frequency release. Both low- and high-frequency stimulated release were almost completely blocked by removal of calcium, indicating activity dependence. Reducing dopamine efflux did not affect adenosine release but inhibiting ionotropic glutamate receptors did, indicating that adenosine release is dependent on downstream effects of glutamate. Therefore, adenosine release after short, high-frequency physiological stimulations is independent of transporter activity or ATP metabolism, and may be due to direct release of adenosine after glutamate receptor activation.
Collapse
Affiliation(s)
- Megan L. Pajski
- Chemistry Department, University of Virginia, P.O. Box 400319, Charlottesville, Virginia 22904, United States
| | - B. Jill Venton
- Chemistry Department, University of Virginia, P.O. Box 400319, Charlottesville, Virginia 22904, United States
| |
Collapse
|
13
|
Adenosine A1 receptor agonists reduce hyperalgesia after spinal cord injury in rats. Spinal Cord 2010; 48:685-90. [PMID: 20065990 DOI: 10.1038/sc.2009.194] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN An in vivo study using a spinal cord compression model in rats. OBJECTIVES To evaluate the effect of adenosine on thermal hyperalgesia after spinal cord injury (SCI). SUMMARY OF BACKGROUND DATA After SCI, some patients suffer dysesthesia that is unresponsive to conventional treatments. We previously established a rat thoracic spinal cord mild-compression model by which we were able to induce thermal hyperalgesia in the hind limbs. METHODS The thoracic spinal cord was compressed gently using a 20-g weight for 20 min. The withdrawal latency in response to thermal stimulation was monitored bilaterally in the hind limbs using Hargreaves' Plantar test apparatus. RESULTS SCI-induced thermal hyperalgesia was mimicked by the intrathecal application of 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), a selective adenosine A1 receptor antagonist. Hyperalgesia induced by SCI was significantly inhibited by the intrathecal application of 10-30 nmol chloro-adenosine (Cl-adenosine), a nonselective adenosine receptor agonist. The effect of Cl-adenosine (10 nmol) on hyperalgesia after SCI was blocked by the simultaneous application of DPCPX. Intrathecal application of R(-)N6-(2phenylisopropyl) adenosine (R-PIA; 10 nmol), a selective A1 receptor agonist, also inhibited SCI-induced hyperalgesia. In contrast, intrathecal application of CGS21680, a selective adenosine A2a receptor agonist, did not inhibit SCI-induced hyperalgesia. CONCLUSIONS These results suggest that adenosine inhibits hyperalgesia through the stimulation of A1 receptors. Adenosine or adenosine A1 receptor agonists should be considered as candidates for new therapeutic methods for treating post-SCI dysesthesia.
Collapse
|
14
|
Liu DZ, Zhu J, Jin DZ, Zhang LM, Ji XQ, Ye Y, Tang CP, Zhu XZ. Behavioral recovery following sub-chronic paeoniflorin administration in the striatal 6-OHDA lesion rodent model of Parkinson's disease. JOURNAL OF ETHNOPHARMACOLOGY 2007; 112:327-32. [PMID: 17451897 DOI: 10.1016/j.jep.2007.03.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 03/04/2007] [Accepted: 03/13/2007] [Indexed: 05/15/2023]
Abstract
In the present studies, the effect of paeoniflorin (PF), one of the main compounds extracted from Paeoniae radix, in alleviating the neurological impairment following unilateral striatal 6-hydroxydopamine (6-OHDA) lesion was examined in Sprague-Dawley rats. Sub-chronic PF (2.5, 5 and 10 mg/kg, s.c., twice daily for 11 days) administration dose-dependently reduced apomorphine (APO)-induced rotation, suggesting that PF had an ameliorative effect on the 6-OHDA-induced neurological impairment. Notably, PF had no direct action on dopamine D(1) receptor or dopamine D(2) receptor indicated by the competitive binding experiments. These results suggest that PF, an active component of Paeoniae radix, might provide an opportunity to introduce a non-dopaminergic management of Parkinson's disease.
Collapse
Affiliation(s)
- Da-Zhi Liu
- Department of Pharmacology II, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Liu DZ, Xie KQ, Ji XQ, Ye Y, Jiang CL, Zhu XZ. Neuroprotective effect of paeoniflorin on cerebral ischemic rat by activating adenosine A1 receptor in a manner different from its classical agonists. Br J Pharmacol 2006; 146:604-11. [PMID: 16086036 PMCID: PMC1751175 DOI: 10.1038/sj.bjp.0706335] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The effects of paeoniflorin (PF), a compound isolated from Paeony radix, on neurological impairment and histologically measured infarction volume following transient and permanent focal ischemia were examined in Sprague-Dawley rats. In transient ischemia model, rats were subjected to a 1.5-h occlusion of the middle cerebral artery (MCA). The administration of PF (2.5 and 5 mg kg(-1), s.c.) produced a dose-dependent decrease in both neurological impairment and the histologically measured infarction volume. Similar results were also obtained when PF (2.5, 5, and 10 mg kg(-1), s.c.) was given in permanent ischemia model. The neuroprotective effect of PF (10 mg kg(-1), s.c.) was abolished by pretreatment of DPCPX (0.25 mg kg(-1), s.c.), a selective adenosine A1 receptor (A1R) antagonist. PF (10, 40, and 160 mg kg(-1), i.v.) had no effect on mean arterial pressure (MAP) and heart rates (HR) in the conscious rat. Additionally, PF (10(-3) mol l(-1)) had no effect on noradrenaline- (NA-) or high K+ concentration-induced contractions of isolated rabbit primary artery. In competitive binding experiments, PF did not compete with the binding of [3H]DPCPX, but displaced the binding of [3H]NECA to the membrane preparation of rat cerebral cortex. This binding manner was distinguished from the classical A1R agonists. The results demonstrated that activation of A1R might be involved in PF-induced neuroprotection in cerebral ischemia in rat. However, PF had no 'well-known' cardiovascular side effects of classical A1R agonists. The results suggest that PF might have the potential therapeutic value as an anti-stroke drug.
Collapse
MESH Headings
- Adenosine-5'-(N-ethylcarboxamide)/metabolism
- Animals
- Benzoates/administration & dosage
- Benzoates/metabolism
- Benzoates/pharmacology
- Binding, Competitive
- Bridged-Ring Compounds/administration & dosage
- Bridged-Ring Compounds/metabolism
- Bridged-Ring Compounds/pharmacology
- Cerebral Cortex/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Glucosides/administration & dosage
- Glucosides/metabolism
- Glucosides/pharmacology
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/prevention & control
- Inhibitory Concentration 50
- Ischemic Attack, Transient/pathology
- Ischemic Attack, Transient/prevention & control
- Male
- Monoterpenes
- Neuroprotective Agents/administration & dosage
- Neuroprotective Agents/metabolism
- Neuroprotective Agents/pharmacology
- Paeonia
- Plant Roots
- Rats
- Rats, Sprague-Dawley
- Receptor, Adenosine A1/drug effects
- Receptor, Adenosine A1/metabolism
- Time Factors
- Xanthines/administration & dosage
- Xanthines/pharmacology
Collapse
Affiliation(s)
- Da-Zhi Liu
- Department of Pharmacology, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, China
| | - Ke-Qiang Xie
- Department of Pharmacology, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, China
| | - Xin-Quan Ji
- Department of Pharmacology, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, China
| | - Yang Ye
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, China
| | - Cheng-Liang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, China
| | - Xing-Zu Zhu
- Department of Pharmacology, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, China
- Author for correspondence:
| |
Collapse
|
16
|
Yates L, Clark JH, Martin TJ, James S, Broadley KJ, Kidd EJ. Radioligand binding and functional responses of ligands for human recombinant adenosine A(3) receptors. ACTA ACUST UNITED AC 2006; 26:191-200. [PMID: 16553647 DOI: 10.1111/j.1474-8673.2006.00372.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The binding and functional properties of adenosine receptor ligands were compared in Chinese hamster ovary cells transfected with human adenosine A(3) receptors. Inhibition of [(125)I]-aminobenzyl-5'-N-methylcarboamidoadenosine ([(125)I]-AB-MECA) binding by adenosine receptor ligands was examined in membrane preparations. Inhibition of forskolin-induced cAMP accumulation by agonists was measured using a cAMP enzyme immunoassay. The rank order of agonist potency for both assays was N(6)-(3-iodobenzyl)-adenosine-5'-N-methyluronamide (IB-MECA) > 5'-N-ethylcarboxamidoadenosine (NECA) > (-)-N(6)-[(R)-phenylisopropyl] adenosine (R-PIA) > 4-aminobenzyl-5'-N-methylcarboxamidoadenosine (AB-MECA) > N(6)-cyclopentyl adenosine (CPA) > adenosine. The radioligand binding rank order of antagonist potency was N-[9-chloro-2-(2-furanyl)[1,2,4]-triazolo[1,5-c]quinazolin-5-benzeneacetamide (MRS1220) > 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) > 8-phenyltheophylline (8-PT) > 8-(p-sulfophenyl)-theophylline (8-SPT). MRS1220 competitively inhibited the effect of IB-MECA on cAMP production, with a K(B) value of 0.35 nm. These data are characteristic of adenosine A(3) receptors. The absence of Mg(2+) and presence of guanosine 5'-(gamma-thio)triphosphate (GTPgammaS) significantly reduced agonist binding inhibition potency, indicating binding to high- and low-affinity states. The IB-MECA, NECA and R-PIA IC(50) values were greater for the cAMP assay than for radioligand binding, suggesting an efficient stimulus-response transduction pathway.
Collapse
Affiliation(s)
- L Yates
- Division of Pharmacology, Welsh School of Pharmacy, Cardiff University, King Edward VII Avenue, Cardiff CF10 3XF, UK
| | | | | | | | | | | |
Collapse
|
17
|
Marro PJ, Mishra OP, Delivoria-Papadopoulos M. Effect of allopurinol on brain adenosine levels during hypoxia in newborn piglets. Brain Res 2006; 1073-1074:444-50. [PMID: 16443203 DOI: 10.1016/j.brainres.2005.11.061] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 11/11/2005] [Accepted: 11/12/2005] [Indexed: 10/25/2022]
Abstract
Adenosine, a purine nucleoside, is a potent inhibitory neuromodulator in the brain which may provide an important endogenous neuroprotective role during hypoxia-ischemia. Allopurinol, a xanthine oxidase inhibitor, blocks purine degradation and may result in the accumulation of purine metabolites, including adenosine, during hypoxia. The present study determines the effect of allopurinol administration prior to hypoxia on brain levels of adenosine and purine metabolites in the newborn piglet. Twenty-two newborn piglets (age 3-7 days) were studied: 5 untreated normoxic and 6 allopurinol-treated normoxic controls were compared to 5 untreated hypoxic and 6 allopurinol-treated hypoxic animals. Brain tissue energy metabolism was continuously monitored during hypoxia by (31)P NMR spectroscopy. Brain tissue levels of purines increased in both hypoxic groups during hypoxia, however, there were significantly higher increases in brain tissue levels of adenosine (66.5 +/- 30.5 vs. 19.4 +/- 10.7 nmol/gm), P < 0.01 and inosine (265 +/- 97.6 vs. 162.8 +/- 38.3 nmol/gm), P = 0.05 in the allopurinol-treated hypoxic group. Allopurinol inhibits purine degradation under severe hypoxic conditions and results in a significant increase in brain tissue levels of adenosine and inosine. The increased accumulation of CNS adenosine during hypoxia which is seen in the allopurinol-treated animals may potentiate adenosine's intrinsic neuroprotective mechanisms.
Collapse
Affiliation(s)
- Peter J Marro
- Department of Pediatrics, Division of Neonatology, Barbara Bush Children's Hospital at Maine Medical Center, 22 Bramhall Street, Portland, ME 04102-3134, USA.
| | | | | |
Collapse
|
18
|
Lai DM, Tu YK, Liu IM, Cheng JT. Increase of adenosine A1 receptor gene expression in cerebral ischemia of Wistar rats. Neurosci Lett 2005; 387:59-61. [PMID: 16055264 DOI: 10.1016/j.neulet.2005.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2005] [Revised: 07/08/2005] [Accepted: 07/11/2005] [Indexed: 10/25/2022]
Abstract
In an attempt to know the role of adenosine A1 receptor in cerebral ischemia, the present study employed the ligation of bilateral carotid arteries to induce ischemia in Wistar rats. Changes of gene expression of adenosine A1 receptor in cerebral cortex of ischemic rats were compared with normal sham control and reperfusion group that received regular blood flow after a transient ischemia. The mRNA level of adenosine A1 receptor in cerebral cortex was markedly raised by this artificial ischemia. Also, reperfusion reversed this elevation to a level near the control. This change was also observed at the protein level using Western blot analysis of adenosine A1 receptor. The raised protein level of adenosine A1 receptor by ischemia was reversed to normal level after reperfusion. These data suggest that the gene expression of adenosine A1 receptor was increased by ischemia probably due to the compensative response of brain. The raised adenosine A1 receptor may play a protective role in these damaged tissues.
Collapse
Affiliation(s)
- Dar-Ming Lai
- Neurosurgical Division, Department of Surgery, National Taiwan University Hospital, Taipei 10001, Taiwan
| | | | | | | |
Collapse
|
19
|
Chauhan NB, Siegel GJ, Feinstein DL. Propentofylline attenuates tau hyperphosphorylation in Alzheimer's Swedish mutant model Tg2576. Neuropharmacology 2005; 48:93-104. [PMID: 15617731 DOI: 10.1016/j.neuropharm.2004.09.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2004] [Revised: 08/23/2004] [Accepted: 09/02/2004] [Indexed: 01/05/2023]
Abstract
Key pathological hallmarks of Alzheimer's disease (AD) are the deposition of amyloid plaques containing Abeta-peptides and the formation of neurofibrillary tangles containing hyperphosphorylated tau. Propentofylline (PPF) is a synthetic xanthine derivative that inhibits phosphodiesterase and adenosine uptake. These effects of PPF influence many cellular functions including stimulating synthesis/release of nerve growth factor. We tested the effects of PPF on disease progression in transgenic mice overexpressing the Swedish mutant human APP (Tg2576). The untreated Tg mice show, together with increased amyloidogenesis, increased levels of tau hyperphosphorylation and increased ratios of the activated to inactivated GSK-3beta, one of the key kinases that can phosphorylate tau. One month of PPF feeding (40 mg/kg per day) reduced the burden of amyloid plaques and the levels of hyperphosphorylated tau and immunoreactive IL-1beta. In parallel with these changes, PPF reduced the activated form of GSK-3beta and increased the inactivated form of GSK-3beta, restoring their ratio almost to normal values. These results demonstrate that PPF can exert multiple protective effects on both amyloidogenesis and tau hyperphosphorylation in an animal model of AD. Our earlier report [Neurochem. Int. 43(3) (2003) 225] demonstrated that Tg2576 animals show decreased levels of mRNA for NGF with increased amyloid burden while feeding of PPF results in a major shift from beta-amyloidogenic to alpha-secretory processing of APP together with increased expression of NGF mRNA. The current new data enlarge our understanding of PPF effects in brain and of tau hyperphosphorylation in Tg animals and are consistent with the hypothesis that GSK-3beta is a nodal point linking amyloid and tau pathology. Therapeutic interventions directed toward multiple pathological processes may be more protective than treatments directed toward a single process. The new results reported here indicate that further testing of PPF as a potential therapy in AD is warranted.
Collapse
Affiliation(s)
- Neelima B Chauhan
- Research and Development (151), VA Chicago Health Care System West Side Division, Jesse Brown VA Medical Center Chicago, 820 South Damen Avenue, Chicago, IL 60612, USA.
| | | | | |
Collapse
|
20
|
Alloisio S, Cugnoli C, Ferroni S, Nobile M. Differential modulation of ATP-induced calcium signalling by A1 and A2 adenosine receptors in cultured cortical astrocytes. Br J Pharmacol 2004; 141:935-42. [PMID: 14993103 PMCID: PMC1574277 DOI: 10.1038/sj.bjp.0705707] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2003] [Revised: 01/14/2004] [Accepted: 01/20/2004] [Indexed: 11/09/2022] Open
Abstract
1. Despite the accumulating evidence that under various pathological conditions the extracellular elevation of adenine-based nucleotides and nucleosides plays a key role in the control of astroglial reactivity, how these signalling molecules interact in the regulation of astrocyte function is still largely elusive. 2. The action of the nucleoside adenosine in the modulation of the intracellular calcium signalling ([Ca(2+)](i)) elicited by adenosine 5'-triphosphate (ATP)-induced activation of P2 purinoceptors was investigated on neocortical type-1 astrocytes in primary culture by using single-cell microfluorimetry. 3. Astrocyte challenge with ATP (1-10 microm) elicited biphasic [Ca(2+)](i) responses consisting of an initial peak followed by a sustained elevation. The stable adenosine analogue 2-chloroadenosine (2-ClA) potentiated the transient [Ca(2+)](i) rise induced by activation of metabotropic P2Y receptors. Among the various P1 receptor agonists tested, the nonselective agonist 5'-N-ethylcarboxamidoadenosine (NECA) mimicked the 2-ClA action, whereas the selective A1 R(-) N6-(2-phenylisopropyl)-adenosine (R-PIA), the A2A 2-[4-(2-carboxyethyl)phenethylamino]-5'-N-ethylcarboxamidoadenosine (CGS-21680) and A3 1-deoxy-1-(6-[([3-lodophenyl]methyl)-amino]-9H-purin-9-yl)-N-methyl-beta-d-ribofuranuronamide (IB-MECA) agonists were ineffective. 4. Application of R-PIA>NECA>or=2-ClA depressed the [Ca(2+)](i) plateau reversibly. Moreover, in the presence of R-PIA or 2-ClA, the prolonged [Ca(2+)](i) signal was maintained by application of the A1 antagonist 1,3-diethyl-8-phenylxanthine (DPX). Finally, preincubation of the astrocytes with pertussis toxin abrogated the 2-ClA inhibition of the ATP-elicited sustained [Ca(2+)](i) rise without affecting the transient [Ca(2+)](i) potentiation. 5. Taken together, these findings indicate that stimulation of A1 and A2 adenosine receptors mediates a differential modulation of [Ca(2+)](i) signalling elicited by P2 purinoceptors. Since variations in [Ca(2+)](i) dynamics also affect cell proliferation and differentiation, our data suggest that tuning of the extracellular levels of adenosine may be relevant for the control of astrogliosis mediated by adenine nucleotides.
Collapse
Affiliation(s)
- Susanna Alloisio
- Institute of Biophysics, CNR, Via De Marini 6, 16149 Genoa, Italy
| | - Carlo Cugnoli
- Institute of Biophysics, CNR, Via De Marini 6, 16149 Genoa, Italy
| | - Stefano Ferroni
- Department of Human and General Physiology, University of Bologna, Via San Donato, 19-2–40127 Bologna, Italy
| | - Mario Nobile
- Institute of Biophysics, CNR, Via De Marini 6, 16149 Genoa, Italy
| |
Collapse
|
21
|
Blum D, Hourez R, Galas MC, Popoli P, Schiffmann SN. Adenosine receptors and Huntington's disease: implications for pathogenesis and therapeutics. Lancet Neurol 2003; 2:366-74. [PMID: 12849153 DOI: 10.1016/s1474-4422(03)00411-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Huntington's disease (HD) is a devastating hereditary neurodegenerative disorder, the progression of which cannot be prevented by any neuroprotective approach, despite major advances in the understanding of its pathogenesis. The study of several animal models of the disease has led to the discovery of both loss-of-normal and gain-of-toxic functions of the mutated huntingtin protein and the elucidation of the mechanisms that underlie the formation of huntingtin aggregates and nuclear inclusions. Moreover, these models also provide good evidence of a role for excitotoxicity and mitochondrial metabolic impairments in striatal neuronal death. Adenosine has neuroprotective potential in both acute and chronic neurological disorders such as stroke or Parkinson's disease. Here we review experimental data on the role of A1 and A2A adenosine receptors in HD that warrant further investigation of the beneficial effects of A1 agonists and A2A antagonists in animal models of HD. Future pharmacological analysis of adenosine receptors could justify the use of A1 agonists and A2A antagonists for the treatment of HDin clinical trials.
Collapse
Affiliation(s)
- David Blum
- Laboratory of Neurophysiology, Université Libre de Bruxelles, Belgium.
| | | | | | | | | |
Collapse
|
22
|
Koriyama Y, Chiba K, Mohri T. Propentofylline protects beta-amyloid protein-induced apoptosis in cultured rat hippocampal neurons. Eur J Pharmacol 2003; 458:235-41. [PMID: 12504778 DOI: 10.1016/s0014-2999(02)02789-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
beta-Amyloid protein 1-42 (beta42) can induce apoptosis in the cultured hippocampal neurons, suggesting that it plays an important role in causing neurodegeneration in Alzheimer's disease. Recently, propentofylline, a synthetic xanthine derivative, has been reported to depress ischemic degeneration of hippocampal neurons in gerbils. The present study investigated whether or not propentofylline affected the beta42-induced apoptosis of hippocampal neurons, and if so, which type of signaling machinery works in the neuroprotective action of propentofylline. Addition of propentofylline markedly attenuated the beta42-induced cell death of rat hippocampal neurons. The amyloid protein certainly induced apoptosis in the cultured hippocampal cells revealed by nuclear condensation, caspase-3 activation and an increase of Bax. Intriguingly, propentofylline blocked both the apoptotic features induced by beta42 and further induced an anti-apoptotic protein, Bcl-2, during a short time of incubation. The neuroprotective action of propentofylline was comparably replaced with dibutyryl cAMP (dbcAMP) and was completely suppressed by a low concentration of specific protein kinase A (PKA) inhibitor. Taken altogether, the data strongly suggest that the protection of propentofylline on the beta42-induced neurotoxicity is caused by enhancing anti-apoptotic action through cAMP-PKA system. Propentofylline as a therapeutic agent to Alzheimer's disease is discussed.
Collapse
Affiliation(s)
- Yoshiki Koriyama
- Department of Biodynamics, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Ishikawa, Japan.
| | | | | |
Collapse
|
23
|
Gourine AV, Llaudet E, Thomas T, Dale N, Spyer KM. Adenosine release in nucleus tractus solitarii does not appear to mediate hypoxia-induced respiratory depression in rats. J Physiol 2002; 544:161-70. [PMID: 12356889 PMCID: PMC2290570 DOI: 10.1113/jphysiol.2002.024174] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2002] [Accepted: 06/18/2002] [Indexed: 11/08/2022] Open
Abstract
The time course of adenosine release in the nucleus tractus solitarii (NTS) and ventrolateral medulla (VLM) during acute systemic hypoxia was investigated in the anaesthetised rat by means of amperometric enzymatic sensors. It was found that acute hypoxia induced a significant delayed increase in adenosine level (reaching levels as high as 5 microM) in the NTS and that hypoxia-induced release of adenosine was similar at various regions of the NTS along its rostro-caudal axis. Significantly smaller or no increases in adenosine levels at all in response to hypoxia were observed in the VLM. The increase in adenosine level in the NTS occurred during reoxygenation after the termination of the hypoxic challenge and was accompanied by a smaller increase in inosine concentration. At the dorsal surface of the brainstem, only release of inosine was detected following acute hypoxia. Addition of the ecto-5'-nucleotidase inhibitor alpha,beta-methylene ADP (200 microM) to the dorsal surface of the brainstem completely abolished the signal evoked by hypoxia, suggesting that the inosine arose from adenosine that was produced in the extracellular space by the prior release of ATP. This study indicates that following systemic hypoxia, adenosine levels in the NTS increase to a significantly greater extent than in the VLM. However, the increase in adenosine concentration in the NTS occurs too late to be responsible for the hypoxia-induced depression of the respiratory activity.
Collapse
Affiliation(s)
- Alexander V Gourine
- Department of Physiology, Royal Free and University College London Medical School, Rowland Hill Street, UK.
| | | | | | | | | |
Collapse
|
24
|
Delicado EG, Jiménez AI, Castro E, Miras-Portugal MT. Cerebellar astrocytes coexpress different purinoceptors: Cross-talk between several transduction mechanisms. Drug Dev Res 2001. [DOI: 10.1002/ddr.1105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
25
|
Abstract
Agonist stimulation of adenosine A(1) receptors has been consistently shown to result in reduction of brain damage following experimentally induced global and focal brain ischaemia in animals. Unsurprisingly, the use of adenosine A(1) receptors as targets for the development of clinical therapeutics suitable for treatment of ischaemic brain disorders has been suggested by many authors. The latest studies of adenosine and its receptors indicate that adenosine-mediated actions might be far more complex than originally anticipated, casting some doubt about the rapid development of stroke treatment based on adenosine. This review discusses the possible role of adenosine receptor subtypes (A(1), A(2) and A(3)) in the context of their potential as therapeutics in stroke.
Collapse
Affiliation(s)
- D K von Lubitz
- Emergency Medicine Research Laboratories, Department of Emergency Medicine, University of Michigan Medical Center, TC/B1354/0303, 1500 E. Medical Center Drive, Ann Arbor, MI 48109-0303, USA.
| |
Collapse
|
26
|
Alves PM, Fonseca LL, Peixoto CC, Almeida AC, Carrondo MJ, Santos H. NMR studies on energy metabolism of immobilized primary neurons and astrocytes during hypoxia, ischemia and hypoglycemia. NMR IN BIOMEDICINE 2000; 13:438-448. [PMID: 11252029 DOI: 10.1002/nbm.665] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Changes in high-energy phosphate metabolites (ATP and phosphocreatine) were monitored, in real time, by 31P-nuclear magnetic resonance in primary cell cultures of neurons and astrocytes during periods of hypoxia, ischemia and hypoglycemia, and also during the recovery periods following the re-establishment of standard conditions. Cells were immobilized in basement membrane gel threads and perfused with oxygen-depleted medium (oxygen concentration below 30 microM), to create hypoxic conditions, or with aerobic medium (oxygen concentration approximately 460 microM) containing different concentrations of glucose (hypoglycemia). Ischemic conditions were imposed by stopping perfusion for different periods of time (15 min to 2 h). The experimental set-up enabled the acquisition of 31P-spectra with high signal-to-noise ratio within 10-20 min for both cell types. The effect of hypoxia on glucose metabolism was assessed by 13C-NMR using [1-13C]glucose as substrate. The levels of ATP and PCr in astrocytes were unaffected during hypoxia (up to 2 h), but decreased notably under ischemia. In neurons, hypoxic periods caused a sharp drop of the ATP and PCr levels, and considerable damage to the capacity of neurons to replenish the ATP and PCr pools upon returning to normoxic conditions. However, neurons were remarkably less sensitive to ischemic conditions, the ATP and PCr pools being restored quickly, even after 2 h under challenging conditions. The data show that neurons were more resistant to ischemia than astrocytes, and suggest that the capacity to sustain the pools of ATP and PCr was part of the neuronal protective strategy.
Collapse
Affiliation(s)
- P M Alves
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | | | | | | | | |
Collapse
|
27
|
Sinclair CJ, LaRivière CG, Young JD, Cass CE, Baldwin SA, Parkinson FE. Purine uptake and release in rat C6 glioma cells: nucleoside transport and purine metabolism under ATP-depleting conditions. J Neurochem 2000; 75:1528-38. [PMID: 10987833 DOI: 10.1046/j.1471-4159.2000.0751528.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Adenosine, through activation of membrane-bound receptors, has been reported to have neuroprotective properties during strokes or seizures. The role of astrocytes in regulating brain interstitial adenosine levels has not been clearly defined. We have determined the nucleoside transporters present in rat C6 glioma cells. RT-PCR analysis, (3)H-nucleoside uptake experiments, and [(3)H]nitrobenzylthioinosine ([(3)H]NBMPR) binding assays indicated that the primary functional nucleoside transporter in C6 cells was rENT2, an equilibrative nucleoside transporter (ENT) that is relatively insensitive to inhibition by NBMPR. [(3)H]Formycin B, a poorly metabolized nucleoside analogue, was used to investigate nucleoside release processes, and rENT2 transporters mediated [(3)H]formycin B release from these cells. Adenosine release was investigated by first loading cells with [(3)H]adenine to label adenine nucleotide pools. Tritium release was initiated by inhibiting glycolytic and oxidative ATP generation and thus depleting ATP levels. Our results indicate that during ATP-depleting conditions, AMP catabolism progressed via the reactions AMP --> IMP --> inosine --> hypoxanthine, which accounted for >90% of the evoked tritium release. It was surprising that adenosine was not released during ATP-depleting conditions unless AMP deaminase and adenosine deaminase were inhibited. Inosine release was enhanced by inhibition of purine nucleoside phosphorylase; ENT2 transporters mediated the release of adenosine or inosine. However, inhibition of AMP deaminase/adenosine deaminase or purine nucleoside phosphorylase during ATP depletion produced release of adenosine or inosine, respectively, via the rENT2 transporter. This indicates that C6 glioma cells possess primarily rENT2 nucleoside transporters that function in adenosine uptake but that intracellular metabolism prevents the release of adenosine from these cells even during ATP-depleting conditions.
Collapse
Affiliation(s)
- C J Sinclair
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | |
Collapse
|
28
|
de Mendonça A, Sebastião AM, Ribeiro JA. Adenosine: does it have a neuroprotective role after all? BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2000; 33:258-74. [PMID: 11011069 DOI: 10.1016/s0165-0173(00)00033-3] [Citation(s) in RCA: 200] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A neuroprotective role for adenosine is commonly assumed. Recent studies revealed that adenosine may unexpectedly, under certain circumstances, have the opposite effects contributing to neuronal damage and death. The basis for this duality may be the activation of distinct subtypes of adenosine receptors, interactions between these receptors, differential actions on neuronal and glial cells, and various time frames of adenosinergic compounds administration. If these aspects are understood, adenosine should remain an interesting target for therapeutical neuroprotective approaches after all.
Collapse
Affiliation(s)
- A de Mendonça
- Laboratory of Neurosciences, Faculty of Medicine of Lisbon, Av. Professor Egas Moniz, 1649-035, Lisbon, Portugal.
| | | | | |
Collapse
|
29
|
Reshef A, Sperling O, Zoref-Shani E. The adenosine-induced mechanism for the acquisition of ischemic tolerance in primary rat neuronal cultures. Pharmacol Ther 2000; 87:151-9. [PMID: 11007997 DOI: 10.1016/s0163-7258(00)00045-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neurons can be preconditioned by various procedures to resist ischemic insult. The preconditioning mechanism induced by adenosine ("the adenosine mechanism") was characterized in primary rat neuronal cultures, employing a model of chemical ischemia. The protective mechanism, initiated by activation of adenosine receptors, consists of a signal transduction pathway, involving activation of protein kinase C (PKC) and opening of ATP-sensitive potassium (K(ATP)) channels. Direct activation (and inhibition) of PKC, as well as opening of K(ATP) channels, also confers protection. The opening of the K(ATP) channels mediates the signal activated by the adenosine receptors, and probably also that activated by PKC. The acquired ischemic resistance lasts up to 5 days, depending on the activating substance. The adenosine-activated cascade of events leading to ischemic tolerance in neurons is similar to that operating in cardiomyocytes.
Collapse
Affiliation(s)
- A Reshef
- Department of Clinical Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | |
Collapse
|
30
|
Martinez-Tica JF, Zornow MH. Effects of adenosine agonists and an antagonist on excitatory transmitter release from the ischemic rabbit hippocampus. Brain Res 2000; 872:110-5. [PMID: 10924682 DOI: 10.1016/s0006-8993(00)02483-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The purpose of this study was to determine the effects of adenosine agonists and an antagonist on ischemia-induced extracellular glutamate concentrations in an animal model of transient cerebral ischemia using in vivo cerebral microdialysis. Fifty New Zealand white rabbits were randomly assigned to one of five groups (normothermia, hypothermia, cyclopentyladenosine (CPA), theophylline, or propentofylline). Microdialysis probes were stereotactically placed in the dorsal hippocampus. Twenty minutes before the onset of ischemia, either 1 mg/kg CPA, 5 mg/kg propentofylline, or 20 mg/kg theophylline were administered intravenously. Esophageal temperature was maintained at 38 degrees C, except in the hypothermic animals, which were cooled to 30 degrees C throughout the entire experiment. Two 12-min periods of cerebral ischemia, separated by a 105-min interval of reperfusion, were produced by inflating a neck tourniquet. High-performance liquid chromatography was used to determine the glutamate concentration in the microdialysate. There were no significant increases in glutamate concentrations during the first ischemic period in any of the five groups. During the second ischemic episode, glutamate concentrations in the normothermic group peaked at levels approximately three times higher than the initial values. A similar pattern of changes in glutamate concentrations was observed in the CPA, propentofylline, and theophylline groups. In the hypothermic group, the concentrations of glutamate remained at baseline levels during the entire experiment. Contrary to expectations, neither the adenosine agonists (CPA, propentofylline) nor the antagonist (theophylline) had any effect on extracellular glutamate concentrations in the peri-ischemic period. Although adenosine and its analogs may be cerebroprotective agents, their mechanism of action is not fully understood. The data derived from this study indicates that the acute administration of such agents had no effect on ischemia-induced glutamate release within the hippocampus under these experimental conditions. Based on these results, further work is needed to compare in vivo versus in vitro experimental results in acute and long-term treatment studies with adenosine receptor agonists and antagonists.
Collapse
Affiliation(s)
- J F Martinez-Tica
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX 77555-0591, USA
| | | |
Collapse
|
31
|
Dale N, Pearson T, Frenguelli BG. Direct measurement of adenosine release during hypoxia in the CA1 region of the rat hippocampal slice. J Physiol 2000; 526 Pt 1:143-55. [PMID: 10878107 PMCID: PMC2269993 DOI: 10.1111/j.1469-7793.2000.00143.x] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2000] [Accepted: 03/31/2000] [Indexed: 11/28/2022] Open
Abstract
We have used an enzyme-based, twin-barrelled sensor to measure adenosine release during hypoxia in the CA1 region of rat hippocampal slices in conjunction with simultaneous extracellular field recordings of excitatory synaptic transmission. When loaded with a combination of adenosine deaminase, nucleoside phosphorylase and xanthine oxidase, the sensor responded linearly to exogenous adenosine over the concentration range 10 nM to 20 microM. Without enzymes, the sensor when placed on the surface of hippocampal slices recorded a very small net signal during hypoxia of 40 +/- 43 pA (mean +/- s.e.m.; n = 7). Only when one barrel was loaded with the complete sequence of enzymes and the other with the last two in the cascade did the sensor record a large net difference signal during hypoxia (1226 +/- 423 pA; n = 7). This signal increased progressively during the hypoxic episode, scaled with the hypoxic depression of the simultaneously recorded field excitatory postsynaptic potential and was greatly reduced (67 +/- 6.5 %; n = 9) by coformycin (0.5-2 microM), a selective inhibitor of adenosine deaminase, the first enzyme in the enzymic cascade within the sensor. For 5 min hypoxic episodes, the sensor recorded a peak concentration of adenosine of 5.6 +/- 1.2 microM (n = 16) with an IC(50) for the depression of transmission of approximately 3 microM. In slices pre-incubated for 3-6 h in nominally Ca(2+)-free artificial cerebrospinal fluid, 5 min of hypoxia resulted in an approximately 9-fold greater release of adenosine (48.9 +/- 17.7 microM; n = 6). High extracellular Ca(2+) (4 mM) both reduced the adenosine signal recorded by the sensor during hypoxia (3.5 +/- 0.6 microM; n = 4) and delayed the hypoxic depression of excitatory synaptic transmission.
Collapse
Affiliation(s)
- N Dale
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | | |
Collapse
|
32
|
Johnston WE. Preconditioning the Brain and Heart: Implications for Cardiac Surgery. Semin Cardiothorac Vasc Anesth 2000. [DOI: 10.1053/vc.2000.6483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Despite many recent advances in emboli detection, aortic imaging, myocardial preservation, and perfusion equipment, ischemic injury to the heart and brain remains a serious complications after cardiac surgery. Hypoperfusion (particularly in the heart) and microem boli (particularly in the brain) during cardiopulmonary bypass constitute the etiology of ischemia. Although hypothermia has traditionally been the mainstay for systemic protection from transient ischemia, there has been a general trend to accept warmer heart and core temperatures during bypass, which increases the poten tial for ischemic injury to various organs. This article discusses recent advances in the understanding of myocardial and brain preconditioning and their poten tial role to provide additional protection during cardiac surgery.
Collapse
Affiliation(s)
- William E. Johnston
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
33
|
Haag P, Schneider T, Schäbitz W, Hacke W. Effect of propentofylline (HWA 285) on focal ischemia in rats: effect of treatment and posttreatment duration on infarct size. J Neurol Sci 2000; 175:52-6. [PMID: 10785257 DOI: 10.1016/s0022-510x(00)00278-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
UNLABELLED BACKGROUND AND PUROSEe: in this study we tested the potentially neuroprotective properties of propentofylline in a model of focal ischemia with long-term, repeated treatment. METHODS 37 male Wistar rats (280-300 g) underwent permanent occlusion of the middle cerebral artery (MCA). Infusion was started 30 min after occlusion of the MCA over a period of 2 h with a dosage of 0.01 mg/kg body weight. Immediately after the termination of infusion repetitive intraperitoneal injections were started. Animals were assigned to four groups: continuous treatment for a period of 12 h with 24-h survival (group A, n=9) or 48-h survival (group B, n=10), continuous treatment for a period of 48 h with 48-h survival (group C, n=9) and placebo (group D, n=9). Infarct size was calculated from brain slices stained with 2,3,5-triphenyltetrazolium chloride. RESULTS the infarct size was significantly reduced in group C (treatment for 48 h) (163.9+/-30.5 mm(3)) compared to the placebo group (297.4+/-17. 7 mm(3)). No effect on infarct size was observed in group A (196. 8+/-37.3 mm(3)) and group B (239.6+/-42.9 mm(3)) compared to placebo. CONCLUSION continuous i.p. injections of propentofylline over a period of 48 h significantly reduces infarct size in an animal model of focal cerebral ischemia. With shorter periods of continuous administration of the drug and delayed postmortem analysis, reductions in the infarct size did not reach a level of significance. These data show the importance of continuous long-term administration after ischemic stroke in clinical trials to achieve the beneficial effects of neuroprotection by propentofylline.
Collapse
Affiliation(s)
- P Haag
- Department of Neurology, University of Heidelberg, Germany.
| | | | | | | |
Collapse
|
34
|
Koriyama Y, Yamazaki M, Chiba K, Mohri T. Evaluation of neurotoxicity of alzheimer's amyloid beta protein (beta42) in cultured hippocampal cells and its prevention by propentofylline. JAPANESE JOURNAL OF PHARMACOLOGY 2000; 82:301-6. [PMID: 10875749 DOI: 10.1254/jjp.82.301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Neurotoxicity of beta42 (20 microM) in cultured rat hippocampal neurons was evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction and lactate dehydrogenase (LDH) release methods as quantitative assays of cell death, and both methods indicated that propentofylline (PPF) had the ability to protect the neurons against the toxicity, although these two assay methods revealed different mechanisms for the toxic effect of beta42. Promotion of the active exocytotic system of the cells was suggested after treatment with beta42 in the MTT assay and in determination of 9-aminoacridine (AA) excretion from the preloaded cells after 24-h treatment with beta42. The promotion of AA exocytosis was blocked by the addition of PPF (20 microg/ml). The preventive effect of PPF on the neurotoxicity of beta42 has been proposed to be caused by elevation of the intracellular level of cAMP as a result of depression of the hydrolytic activity of cells.
Collapse
Affiliation(s)
- Y Koriyama
- Department of Biodynamics, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Ishikawa, Japan
| | | | | | | |
Collapse
|
35
|
Schubert P, Morino T, Miyazaki H, Ogata T, Nakamura Y, Marchini C, Ferroni S. Cascading glia reactions: a common pathomechanism and its differentiated control by cyclic nucleotide signaling. Ann N Y Acad Sci 2000; 903:24-33. [PMID: 10818485 DOI: 10.1111/j.1749-6632.2000.tb06346.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A pathological glia activation, stimulated by inflammatory proteins, beta-amyloid, or brain ischemia, is discussed as a common pathogenic factor for progressive nerve cell damage in vascular and Alzheimer dementia. A critical point seems to be reached, if the cytokine-controlled microglial upregulation causes a secondary activation of astrocytes which loose the negative feedback control, are forced to give up their physiological buffering function, and may add to neuronal damage by the release of nitric oxide (NO) and by promoting toxic beta-amyloid formation. A strengthening of the cyclic adenosine-5',3'-monophosphate (cAMP) signaling exerted a differential inhibition of the stimulatory cytokines tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 beta (IL-1 beta) released from cultured rat microglia, but maintained the negative feedback signal IL-6; cAMP inhibited also the release of free oxygen radicals (OR) but not of NO. Reinforcement of the NO-induced cyclic guanosine monophosphate (cGMP) increase by blockade of the phosphodiesterase (PDE) subtype-5 with propentofylline counterbalanced the toxic NO action that causes with OR neuronal damage by peroxynitrate formation. In rat cultured astrocytes, a prolonged cAMP elevation favored cell differentiation, the expression of a mature ion channel patter, and an improvement of the extracellular glutamate uptake. Cyclic AMP signaling could be strengthened by PDE blockade and by raising extracellular adenosine, which stimulates A2 receptor-mediated cAMP synthesis. Via an A1 receptor-mediated effect, elevated adenosine was found to overcome a deficient intracellular calcium mobilization resulting from an impaired muscarinic signaling at pathologically decreased acetylcholine concentrations. We suggest that pharmaca, which elevate extracellular adenosine and/or block the degradation of cyclic nucleotides, may be used to counteract glia-related neuronal damage in dementing processes.
Collapse
Affiliation(s)
- P Schubert
- Department of Neuromorphology, Max Planck Institute of Neurobiology, Martinsried, Germany.
| | | | | | | | | | | | | |
Collapse
|
36
|
Robledo P, Ursu G, Mahy N. Effects of adenosine and gamma-aminobutyric acid A receptor antagonists on N-methyl-D-aspartate induced neurotoxicity in the rat hippocampus. Hippocampus 1999; 9:527-33. [PMID: 10560923 DOI: 10.1002/(sici)1098-1063(1999)9:5<527::aid-hipo6>3.0.co;2-u] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This study investigated the modulatory actions of adenosine and gamma-aminobutyric acid (GABA) on several aspects of N-methyl-D-aspartate (NMDA)-induced neurotoxicity, including neuronal loss, atrophy, necrosis, and calcium accumulation in the hippocampus. For this purpose, we combined unilateral intrahippocampal injections of NMDA (24 nmoles) with acute injections of the selective A1 adenosine receptor antagonist DPCPX (0.03 pmoles), the selective adenosine A2a receptor antagonist CSC (1.5 pmoles), a combination of these two antagonists, and injections of the selective GABA A receptor antagonist bicuculline (60 pmoles). Fifteen days after NMDA injection, neuronal loss with preservation of architecture was observed in stratum oriens, pyramidale, radiatum, lacunosum-moleculare, and stratum moleculare of Ammon's horn, and in radial and granular layers of the dentate gyrus. NMDA plus vehicle also produced a small degree of brain tissue necrosis (holes in the structure) in four of five brains. Acute injections of CSC, but not DPCPX or bicuculline, significantly increased the extent of neuronal loss produced by NMDA plus vehicle. CSC in combination with NMDA induced significantly more necrosis than NMDA plus vehicle. A significant degree of atrophy was observed in the hippocampus after treatment with NMDA plus vehicle, and bicuculline significantly increased the magnitude of this atrophy. NMDA-induced calcium deposits were detected within the radiatum and lacunosum-moleculare layers of the hippocampus and in the hilus of the dentate, but not in the stratum oriens, stratum pyramidale, or in the granular layer of the dentate gyrus. However, treatment with the different antagonists did not significantly modify the magnitude of the NMDA-induced calcium deposits. These results reveal a selective vulnerability of certain areas of the hippocampus to the accumulation of calcium deposits, and a selective interaction between adenosine receptors and NMDA-induced neurotoxicity in the hippocampus.
Collapse
Affiliation(s)
- P Robledo
- Depto. de Ciènces Fisiologiques I. Facultad de Medicina, Universidad de Barcelona, Spain
| | | | | |
Collapse
|
37
|
Nakamura Y, Si QS, Kataoka K. Lipopolysaccharide-induced microglial activation in culture: temporal profiles of morphological change and release of cytokines and nitric oxide. Neurosci Res 1999; 35:95-100. [PMID: 10616913 DOI: 10.1016/s0168-0102(99)00071-1] [Citation(s) in RCA: 167] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Microglial activation has been considered as a result of neuronal damage, however, recently it becomes to recognize as a possible cause of the damage in various neurodegenerative diseases. To elucidate the mechanism of the microglial activation, we examined the time course of lipopolysaccharide (LPS)-induced change in morphology and the release of cytokines and nitric oxide (NO) in cultured microglia from neonatal rat brains. With addition of 1 microg/ml LPS, the cell morphology was drastically changed within 3 h from amoeboid shape to bipolar rod shape. The peak time of such morphological change was at 6 h and then returned to small round shape gradually. This transient change in morphology was completely inhibited by 0.1 mM dibutyryl-cAMP. On the other hand, the release of cytokines and NO showed different time courses after stimulation by LPS; at first tumor necrosis factor (TNF)-alpha was released within 1 h lag time, secondly interleukin (IL)-1beta within 3 h, thirdly IL-6, and at last NO was released with about 6 h lag time. The addition of dibutyryl cAMP markedly inhibited the release of TNF-alpha and IL-1beta, but not IL-6 and NO at all. These results suggest that there are at least two different intracellular signaling pathways of LPS-induced microglial activation; one for early release of TNF-alpha and IL-1beta sensitive to dibutyryl-cAMP and the other for late release of IL-6 and NO insensitive to dibutyryl-cAMP. The transient morphological change seems to be associated with the early release based on the sensitivity to dibutyryl-cAMP.
Collapse
Affiliation(s)
- Y Nakamura
- Department of Physiology, Ehime University School of Medicine, Shigenobu, Japan.
| | | | | |
Collapse
|
38
|
Knutsen LJ, Lau J, Petersen H, Thomsen C, Weis JU, Shalmi M, Judge ME, Hansen AJ, Sheardown MJ. N-substituted adenosines as novel neuroprotective A(1) agonists with diminished hypotensive effects. J Med Chem 1999; 42:3463-77. [PMID: 10479279 DOI: 10.1021/jm960682u] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The synthesis and pharmacological profile of a series of neuroprotective adenosine agonists are described. Novel A(1) agonists with potent central nervous system effects and diminished influence on the cardiovascular system are reported and compared to selected reference adenosine agonists. The novel compounds featured are derived structurally from two key lead structures: 2-chloro-N-(1-phenoxy-2-propyl)adenosine (NNC 21-0041, 9) and 2-chloro-N-(1-piperidinyl)adenosine (NNC 90-1515, 4). The agonists are characterized in terms of their in vitro profiles, both binding and functional, and in vivo activity in relevant animal models. Neuroprotective properties assessed after postischemic dosing in a Mongolian gerbil severe temporary forebrain ischemia paradigm, using hippocampal CA1 damage endpoints, and the efficacy of these agonists in an A(1) functional assay show similarities to some reference adenosine agonists. However, the new compounds we describe exhibit diminished cardiovascular effects in both anesthetized and awake rats when compared to reference A(1) agonists such as (R)-phenylisopropyladenosine (R-PIA, 5), N-cyclopentyladenosine (CPA, 2), 4, N-[(1S,trans)-2-hydroxycyclopentyl]adenosine (GR 79236, 26), N-cyclohexyl-2'-O-methyladenosine (SDZ WAG 994, 27), and N-[(2-methylphenyl)methyl]adenosine (Metrifudil, 28). In mouse permanent middle cerebral artery occlusion focal ischemia, 2-chloro-N-[(R)-[(2-benzothiazolyl)thio]-2-propyl]adenosine (NNC 21-0136, 12) exhibited significant neuroprotection at the remarkably low total intraperitoneal dose of 0.1 mg/kg, a dose at which no cardiovascular effects are observed in conscious rats. The novel agonists described inhibit 6, 7-dimethoxy-4-ethyl-beta-carboline-3-carboxylate-induced seizures, and in mouse locomotor activity higher doses are required to reach ED(50) values than for reference A(1) agonists. We conclude that two of the novel adenosine derivatives revealed herein, 12 and 5'-deoxy-5'-chloro-N-[4-(phenylthio)-1-piperidinyl]adenosine (NNC 21-0147, 13), representatives of a new series of P(1) ligands, reinforce the fact that novel selective adenosine A(1) agonists have potential in the treatment of cerebral ischemia in humans.
Collapse
Affiliation(s)
- L J Knutsen
- Health Care Discovery and Development, Novo Nordisk A/S, Novo Nordisk Park, DK-2760 Måløv, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Adenosine, an intercellular messenger that is a product of the metabolism of ATP, plays a major role in neuronal and vascular responses of the retina to alterations in oxygen delivery. Significant changes in adenosine concentration have been measured in the retina during both ischemia and during the subsequent reperfusion period which result in important, but complex, functional effects. Adenosine A1 receptor stimulation produces a protective effect during ischemia, whereas overstimulation of the A2a receptor has deleterious effects. The mechanisms underlying these findings have not been completely determined, but most likely are the result of alterations in excitotoxicity, gene expression, and blood flow. Paradoxically, prolonged increases in adenosine concentration may be injurious to the retina, a consequence of superoxide radical formation secondary to adenosine catabolism. Adenosine is a critical mediator of blood flow changes in response to ischemia. It is a significant component of the retina's compensatory hyperemic response to ischemia, hypoxia, and hypoglycemia. Increasing endogenous adenosine concentrations may be useful in ameliorating post-ischemic hypoperfusion. Overall, current evidence suggests that adenosine is a vital component of the endogenous retinal response to substrate deprivation. Additionally, in vitro studies provide strong evidence that adenosine is a mediator of the formation and effects of vascular endothelial growth factor, which in turn promotes neovascularization. Finally, the ability of the retina to develop an ischemia-tolerant state by ischemic preconditioning is an intriguing phenomenon that reveals yet another essential role for adenosine in the retina's endogenous response to ischemia. The experimental results described in this review suggest that continued investigation into the role of adenosine in the retina may lead to important clinical applications for adenosine-based therapies that could decrease the incidence of retinal damage in ischemic vasculopathies such as diabetes, glaucoma, and retinal vascular occlusion.
Collapse
Affiliation(s)
- G J Ghiardi
- Department of Anesthesia and Critical Care, University of Chicago, IL 60637, USA
| | | | | |
Collapse
|
40
|
Siniscalchi A, Rodi D, Gessi S, Campi F, Borea PA. Early changes in adenosine A1 receptors in cerebral cortex slices submitted to in vitro ischemia. Neurochem Int 1999; 34:517-22. [PMID: 10402227 DOI: 10.1016/s0197-0186(99)00028-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The effects of brain ischemia on the maximum binding capacity (Bmax) and affinity (Kd) of A1 receptors were studied in the rat cerebral cortex, with an in vitro approach. The results were correlated with changes in 3H-adenosine release, studied under identical experimental conditions. Fifteen minutes of in vitro 'ischemia' (hypoxic, glucose-free medium) induced a significant increase in both Bmax (2398+/-132 fmol/mg protein, 151% of the control, P < 0.05) and in Kd (2.43+/-0.12 nM, 161% of the control, P < 0.01). At the same time, an increase in tritium efflux from [3H]-adenosine labeled cerebral cortex slices to 324% of the control was observed. A trend toward normalization was evident 5-15 min after 'reoxygenation' (restoring normal medium), but the binding parameters were still altered after 60 min (Bmax 2110+/-82 fmol/mg protein, Kd 2.26+/-0.14 nM, P < 0.01 vs the corresponding control) as was adenosine release (196% of the control). These findings suggest that the increased availability of adenosine and its receptors may be a defense mechanism against ischemic injury, while the reduced affinity of A1 receptors, possibly due to desensitization, may be a sign of ischemia-induced cellular damage.
Collapse
Affiliation(s)
- A Siniscalchi
- Department of Clinical and Experimental Medicine, University of Ferrara, Italy.
| | | | | | | | | |
Collapse
|
41
|
Abstract
Numerous studies have consistently shown that agonist stimulation of adenosine A1 receptors results in a significant reduction of morbidity and mortality associated with global and focal brain ischemia in animals. Based on these observations, several authors have suggested utilization of adenosine A1 receptors as targets for the development of clinically viable drugs against ischemic brain disorders. Recent advent of adenosine A1 receptor agonists characterized by lowered cardiovascular effects added additional strength to this argument. On the other hand, although cardioprotective, adenosine A3 receptor agonists proved severely cerebrodestructive when administered prior to global ischemia in gerbils. Moreover, stimulation of adenosine A3 receptors appears to reduce the efficacy of some of the neuroprotective actions mediated by adenosine A1 receptors. The review discusses the possible role of adenosine receptor subtypes (A1, A2, and A3) in the context of their involvement in the pathology of cerebral ischemia, and analyzes putative strategies for the development of clinically useful strategies based on adenosine and its receptors. It also stresses the need for further experimental studies before definitive conclusions on the usefulness of the adenosine concept in the treatment of brain ischemia can be made.
Collapse
Affiliation(s)
- D K von Lubitz
- Department of Emergency Medicine, University of Michigan Medical Center, Ann Arbor 48109-0303, USA.
| |
Collapse
|
42
|
Gracy KN, Clarke CL, Meyers MB, Pickel VM. N-methyl-D-aspartate receptor 1 in the caudate-putamen nucleus: ultrastructural localization and co-expression with sorcin, a 22,000 mol. wt calcium binding protein. Neuroscience 1999; 90:107-17. [PMID: 10188938 DOI: 10.1016/s0306-4522(98)00440-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Entry of calcium through N-methyl-D-aspartate-type glutamate receptors in the caudate-putamen nucleus is essential for normal motor activity, but can produce cytotoxicity with continued stimulation and subsequent release of intracellular calcium. To determine potential functional sites for N-methyl-D-aspartate receptor activation in this region, we examined the ultrastructural localization of the R1 subunit of the N-methyl-D-aspartate receptor (NMDAR1) in rat brain. In addition, we comparatively examined the localization of NMDAR1 and sorcin, a 22,000 mol. wt calcium binding protein present in certain striatal neurons and involved in calcium-induced calcium release. NMDAR1-like immunoreactivity was seen at synaptic and non-synaptic sites on neuronal plasma membranes. Of 1514 NMDAR1-labeled profiles, 62% were dendrites and dendritic spines and the remainder were mainly unmyelinated axons and axon terminals. Sorcin-like immunoreactivity was present in 39% of the profiles that contained NMDAR1 labeling, most (533/595) of which were dendrites and dendritic spines. Of 1807 sorcin-labeled profiles, 42% were identified, however, as small processes including spine necks and unmyelinated axons or axon terminals. These profiles also occasionally contained NMDAR1 or showed synaptic or appositional contacts with other NMDAR1-immunoreactive neurons. The results of this study suggest that in the caudate-putamen nucleus, activation of NMDA receptors permits calcium influx at plasmalemmal sites mainly on dendrites where sorcin may play a role in calcium-induced calcium release. The presence of sorcin in some, but not all NMDA-containing neurons in the caudate-putamen nucleus has potential implications for the known differential vulnerability of certain striatal neurons to excitotoxins.
Collapse
Affiliation(s)
- K N Gracy
- Department of Neurology and Neuroscience, Cornell University Medical College, New York, NY 10021, USA
| | | | | | | |
Collapse
|
43
|
Jim�nez AI, Castro E, Mirabet M, Franco R, Delicado EG, Miras-Portugal MT. Potentiation of ATP calcium responses by A2B receptor stimulation and other signals coupled to Gs proteins in type-1 cerebellar astrocytes. Glia 1999. [DOI: 10.1002/(sici)1098-1136(199904)26:2<119::aid-glia3>3.0.co;2-d] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
44
|
Mirabet M, Herrera C, Cordero OJ, Mallol J, Lluis C, Franco R. Expression of A2B adenosine receptors in human lymphocytes: their role in T cell activation. J Cell Sci 1999; 112 ( Pt 4):491-502. [PMID: 9914161 DOI: 10.1242/jcs.112.4.491] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Extracellular adenosine has a key role in the development and function of the cells of the immune system. Many of the adenosine actions seem to be mediated by specific surface receptors positively coupled to adenylate cyclase: A2A and A2B. Despite the fact that A2A receptors (A2ARs) can be easily studied due to the availability of the specific agonist CGS21680, a pharmacological and physiological characterization of adenosine A2B receptors (A2BRs) in lymphocytes has not been possible due to the lack of suitable reagents. Here we report the generation and characterization of a polyclonal antipeptide antibody raised against the third extracellular loop of the A2BR human clone which is useful for immunocytochemical studies. This antibody has permitted the detection of A2BR+ cells in lymphocyte samples isolated from human peripheral blood. The pharmacology of cAMP-producing compounds is consistent with the presence of functional A2BRs but not of A2A receptors in these human cells. The percentage of A2BR-expressing cells was similar in the CD4(+) or CD8(+) T cell subpopulations. Interestingly activation signals delivered by either phytohemagglutinin or anti-T cell receptor/CD3 complex antibodies led to a significant increase in both the percentage of cells expressing the receptor and the intensity of the labeling. These receptors are functional since interleukin-2 production in these cells is reduced by NECA but not by R-PIA or CGS21680. These results show that A2BR expression is regulated in T cell activation and suggest that the role of adenosine in lymphocyte deactivation is mediated by A2BRs.
Collapse
Affiliation(s)
- M Mirabet
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, University of Barcelona, Barcelona, Catalonia, Spain.
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Numerous studies have consistently shown that agonist stimulation of adenosine A1 receptors results in a significant reduction of morbidity and mortality associated with global and focal brain ischemia in animals. Based on these observations, several authors have suggested utilization of adenosine A1 receptors as targets for the development of clinically viable drugs against ischemic brain disorders. Recent advent of adenosine A1 receptor agonists characterized by lowered cardiovascular effects added additional strength to this argument. On the other hand, although cardioprotective, adenosine A3 receptor agonists proved severely cerebrodestructive when administered prior to global ischemia in gerbils. Moreover, stimulation of adenosine A3 receptors appears to reduce the efficacy of some of the neuroprotective actions mediated by adenosine A receptors. The review discusses the possible role of adenosine receptor subtypes (A1, A2, and A3) in the context of their involvement in the pathology of cerebral ischemia, and analyzes putative strategies for the development of clinically useful strategies based on adenosine and its receptors. It also stresses the need for further experimental studies before definitive conclusions on the usefulness of the adenosine concept in the treatment of brain ischemia can be made.
Collapse
Affiliation(s)
- D K Von Lubitz
- Department of Emergency Medicine, University of Michigan Medical Center, Ann Arbor 48109-0303, USA.
| |
Collapse
|
46
|
Schubert P, Ogata T, Miyazaki H, Marchini C, Ferroni S, Rudolphi K. Pathological immuno-reactions of glial cells in Alzheimer's disease and possible sites of interference. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 1998; 54:167-74. [PMID: 9850925 DOI: 10.1007/978-3-7091-7508-8_16] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A significant role of a pathological glial cell activation in the pathogenesis of Alzheimer's disease is supported by the growing evidence that inflammatory proteins, which are produced by reactive astrocytes, promote the transformation of diffuse beta-amyloid deposits into the filamentous, neurotoxic form. A number of vicious circles, driven by the release of TNF-a and free oxygen radicals from microglial cells, may cause an upregulated microglial activation and their production of interleukin-1 which triggers, secondarily, the crucial activation of astrocytes. Reactive functional changes of glial cells seem to be controlled by an altered balance of the second messengers Ca2+ and cAMP and can be counterregulated by the endogenous cell modulator adenosine which strengthens the cAMP-dependent signalling chain. A further reinforcement of the homeostatic adenosine effects on glial cells by pharmaca, such as propentofylline, may add to neuroprotection in Alzheimer's disease.
Collapse
Affiliation(s)
- P Schubert
- Department of Neuromorphology, Max Planck Institute for Neurobiology, Martinsried, Federal Republic of Germany
| | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Tatlisumak T, Takano K, Carano RA, Miller LP, Foster AC, Fisher M. Delayed treatment with an adenosine kinase inhibitor, GP683, attenuates infarct size in rats with temporary middle cerebral artery occlusion. Stroke 1998; 29:1952-8. [PMID: 9731623 DOI: 10.1161/01.str.29.9.1952] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Brain ischemia is associated with a marked increase in extracellular adenosine levels. This results in activation of cell surface adenosine receptors and some degree of neuroprotection. Adenosine kinase is a key enzyme controlling adenosine metabolism. Inhibition of this enzyme enhances the levels of endogenous brain adenosine already elevated as a result of the ischemic episode. We studied a novel adenosine kinase inhibitor (AKI), GP683, in a rat focal ischemia model. METHODS Four groups of 10 adult Sprague-Dawley rats were exposed to 90 minutes of temporary middle cerebral artery (MCA) occlusion. Animals were injected intraperitoneally with vehicle, 0.5 mg/kg, 1.0 mg/kg, or 2.0 mg/kg of GP683 30, 150, and 270 minutes after the induction of ischemia by a researcher blinded to treatment group. The animals were euthanatized 24 hours after MCA occlusion, and brains were stained with 2,3,5-triphenyltetrazolium chloride. We measured brain temperatures in a separate group of 6 rats before and after administration of 1.0 mg/kg GP683. RESULTS All treated groups showed a reduction in infarct volumes, but a significant effect was observed only in the 1.0 mg/kg-dose group (44% reduction, P=0.0077). Body weight, physiological parameters, neurological scores, and mortality did not differ among the 4 groups. No apparent behavioral side effects were observed. Brain temperatures did not change after drug injection. CONCLUSIONS Our results indicate that the use of AKIs offers therapeutic potential and may represent a novel approach to the treatment of acute brain ischemia. The therapeutic effect observed was not caused by a decrease in brain temperature.
Collapse
Affiliation(s)
- T Tatlisumak
- Department of Neurology, Helsinki University Central Hospital, Finland.
| | | | | | | | | | | |
Collapse
|
49
|
Plaschke K, Bardenheuer HJ, Weigand MA, Martin E, Hoyer S. Increased ATP production during long-term brain ischemia in rats in the presence of propentofylline. Eur J Pharmacol 1998; 349:33-40. [PMID: 9669493 DOI: 10.1016/s0014-2999(98)00172-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Forty adult rats were subjected to stepwise two- and four-brain vessel occlusion and propentofylline 25 mg/day per kilogram body weight was intraperitoneally administered for 1 week or 3 weeks. Adenosine 5'-triphosphate, creatine phosphate, adenosine 5'-diphosphate and adenosine were determined in rat parietotemporal cortex by high-pressure liquid chromatography; lactate and pyruvate were measured spectrophotometrically. Stepwise and permanent long-term brain vessel occlusion gradually reduced the concentration of energy-rich phosphates and induced a marked increase in the concentration of adenosine, a parameter of ischemia. Three weeks of propentofylline treatment resulted in a significant increase in cerebral adenosine 5'-triphosphate concentration from 2.16 +/- 0.15 [(-)-propentofylline] to 2.70 +/- 0.24 nmol/mg wet weight during four-vessel occlusion (+25%). This was associated with an enhancement of the adenosine 5'-triphosphate/adenosine 5'-diphosphate ratio (+33%), mainly because of the significant reduction in adenosine 5'-diphosphate concentration. Propentofylline did not prevent the increase in lactate concentration during permanent brain vessel occlusion, but significantly reduced the tissue concentration of adenosine. In summary, the results demonstrate that continuous propentofylline administration over 3 weeks induced a striking increase in rat cortical adenosine 5'-triphosphate concentration during long-term brain vessel occlusion. Thus, propentofylline may have possible neuroprotective effects and could be used in the treatment of patients with chronic cerebrovascular disorders.
Collapse
Affiliation(s)
- K Plaschke
- Clinic of Anesthesiology, University of Heidelberg, Germany
| | | | | | | | | |
Collapse
|
50
|
Nakamichi T. [Glutamate neurotoxicity during spinal cord ischemia--the neuroprotective effects of adenosine]. THE JAPANESE JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY : OFFICIAL PUBLICATION OF THE JAPANESE ASSOCIATION FOR THORACIC SURGERY = NIHON KYOBU GEKA GAKKAI ZASSHI 1998; 46:354-60. [PMID: 9619035 DOI: 10.1007/bf03217755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Evidence is accumulating that glutamate, a major neurotransmitter, exerts potent neurotoxic activity during ischemia. In our laboratory, a delayed-onset paraplegia model using rabbits has been developed and described. The severity of the ischemic event in this model, i.e., extracellular glutamate overload, is believed to influence the etiology of this delayed neuronal dysfunction. Adenosine, an endogenous neuromodulator, is released after acute ischemic insult and provides neuroprotection by actions on neuronal and glial cells in the still viable border zone of the ischemic focus. We hypothesized that the neuroprotective action of adenosine is associated with inhibition of glutamate neurotoxicity following ischemia. Infrarenal aortic segments from 11 New Zealand white rabbits were isolated for 5 minutes and infused at a rate of 2 ml/min. Group I (n = 6) received normothermic L-glutamate (20 mM). Group II (n = 5) received 75 mg of adenosine and normothermic L-glutamate (20 mM). Neurologic function was assessed at 6, 24, and 48 hours after surgery according to the modified Tarlov scale, After 48 hours, the rabbits were euthanized and their spinal cords were harvested for histologic examination. The neurologic function of three rabbits in group I showed acute paraplegia and the other three showed delayed-onset paraplegia, whereas all group II animals and nearly intact neurologic function. Histologic examination of spinal cords from rabbits in group I showed evidence of moderate spinal cord injury with central gray matter and adjacent white matter necrosis and axonal swelling, whereas spinal cords from group II revealed no evidence of cord injury. Adenosine A1-receptor activation is suspected to reduce excitatory amino acids by controlling the activation of the voltage-dependent NMDA receptor. These results indicate that the neuroprotective effect of adenosine is associated with inhibition of glutamate neurotoxicity, which initiates a deleterious cascade of biochemical events that ultimately result in delayed-onset paraplegia.
Collapse
Affiliation(s)
- T Nakamichi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|