1
|
Ensandoust T, Khakpour-Taleghani B, Jafari A, Rostampour M, Rohampour K, Ch MH. Effect of simultaneous application of adenosine A1 receptor agonist and A2A receptor antagonist on memory, inflammatory factors, and PSD-95 in lipopolysaccharide-induced memory impairment. Behav Brain Res 2025; 476:115210. [PMID: 39159786 DOI: 10.1016/j.bbr.2024.115210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
The potential role of adenosine, a natural neuroprotective agent, and its receptors in the pathogenesis of Alzheimer's disease has been proposed. The present study aims to examine the effect of administering both an A1 receptor agonist and an A2A adenosine receptor antagonist simultaneously on memory, inflammatory factors, and PSD-95 in an LPS-induced Alzheimer's disease model in rats. Fifty-six male Wistar rats were randomly divided into seven groups: Saline, LPS, Saline + Vehicle, LPS + Vehicle, LPS + SCH58261 (A2A receptor antagonist), LPS + CPA (A1 receptor agonist), LPS + SCH58261+CPA. LPS (3 mg/kg/ip) was used to cause memory impairment. Treatment was performed by intraventricular injection of CPA at a dose of 700 μg and SCH-58261 at 40 μg for ten days. Passive avoidance and Y-maze tests were performed to examine animals' memories. IL-10, TNF-α, and PSD-95 levels were measured in the brain using ELISA and western blot, respectively. Compared to the groups receiving each medication separately, the simultaneous administration of CPA and SCH58261 improved memory (P<0.05). Additionally, compared to the single medication groups, there was a significant increase in IL-10, PSD-95, and a significant decrease in TNF-α in the brain tissue (P<0.05). These findings suggest that the activation of A1 receptors along with A2A receptor inhibition could be a potential therapeutic strategy for Alzheimer's disease. These findings suggest that A1 receptor activation combined with A2A receptor inhibition may be a promising therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Tahereh Ensandoust
- Department of Physiology, School of Medicine, Guilan University of Medical Science, Rasht, Iran
| | | | - Adele Jafari
- Department of Physiology, School of Medicine, Guilan University of Medical Science, Rasht, Iran.
| | - Mohammad Rostampour
- Department of Physiology, School of Medicine, Guilan University of Medical Science, Rasht, Iran; Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Kambiz Rohampour
- Department of Physiology, School of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Mojtaba Hedayati Ch
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
2
|
Karger G, Willker JE, Harders AR, Watermann P, Dringen R. ATP Restoration by ATP-Deprived Cultured Primary Astrocytes. Neurochem Res 2024; 50:13. [PMID: 39549173 PMCID: PMC11569012 DOI: 10.1007/s11064-024-04276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 11/18/2024]
Abstract
A high cellular concentration of adenosine triphosphate (ATP) is essential to fuel many important functions of brain astrocytes. Although cellular ATP depletion has frequently been reported for astrocytes, little is known on the metabolic pathways that contribute to ATP restoration by ATP-depleted astrocytes. Incubation of cultured primary rat astrocytes in glucose-free buffer for 60 min with the mitochondrial uncoupler BAM15 lowered the cellular ATP content by around 70%, the total amount of adenosine phosphates by around 50% and the adenylate energy charge (AEC) from 0.9 to 0.6. Testing for ATP restoration after removal of the uncoupler revealed that the presence of glucose as exclusive substrate allowed the cells to restore within 6 h around 80% of the initial ATP content, while coapplication of adenosine plus glucose enabled the cells to fully restore their initial ATP content within 60 min. A rapid but incomplete and transient ATP restoration was found for astrocytes that had been exposed to adenosine alone. This restoration was completely prevented by application of the pyruvate uptake inhibitor UK5099, the respiratory chain inhibitor antimycin A or by the continuous presence of BAM15. However, the presence of these compounds strongly accelerated the release of lactate from the cells, suggesting that the ribose moiety of adenosine can serve as substrate to fuel some ATP restoration via mitochondrial metabolism. Finally, the adenosine-accelerated ATP restoration in glucose-fed astrocytes was inhibited by the presence of the adenosine kinase inhibitor ABT-702. These data demonstrate that astrocytes require for a rapid and complete ATP restoration the presence of both glucose as substrate and adenosine as AMP precursor.
Collapse
Affiliation(s)
- Gabriele Karger
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Johanna Elisabeth Willker
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Antonia Regina Harders
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Patrick Watermann
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.
- Centre for Environmental Research and Sustainable Technologies, University of Bremen, P.O. Box 330440, 28334, Bremen, Germany.
| |
Collapse
|
3
|
Lemes Dos Santos Sanna P, Bernardes Carvalho L, Cristina Dos Santos Afonso C, de Carvalho K, Aires R, Souza J, Rodrigues Ferreira M, Birbrair A, Martha Bernardi M, Latini A, Foganholi da Silva RA. Adora2A downregulation promotes caffeine neuroprotective effect against LPS-induced neuroinflammation in the hippocampus. Brain Res 2024; 1833:148866. [PMID: 38494098 DOI: 10.1016/j.brainres.2024.148866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Caffeine has been extensively studied in the context of CNS pathologies as many researchers have shown that consuming it reduces pro-inflammatory biomarkers, potentially delaying the progression of neurodegenerative pathologies. Several lines of evidence suggest that adenosine receptors, especially A1 and A2A receptors, are the main targets of its neuroprotective action. We found that caffeine pretreatment 15 min before LPS administration reduced the expression of Il1b in the hippocampus and striatum. The harmful modulation of caffeine-induced inflammatory response involved the downregulation of the expression of A2A receptors, especially in the hippocampus. Caffeine treatment alone promoted the downregulation of the adenosinergic receptor Adora2A; however, this promotion effect was reversed by LPS. Although administering caffeine increased the expression of the enzymes DNA methyltransferases 1 and 3A and decreased the expression of the demethylase enzyme Tet1, this effect was reversed by LPS in the hippocampus of mice that were administered Caffeine + LPS, relative to the basal condition; no significant differences were observed in the methylation status of the promoter regions of adenosine receptors. Finally, the bioinformatics analysis of the expanded network demonstrated the following results: the Adora2B gene connects the extended networks of the adenosine receptors Adora1 and Adora2A; the Mapk3 and Esr1 genes connect the extended Adora1 network; the Mapk4 and Arrb2 genes connect the extended Adora2A network with the extended network of the proinflammatory cytokine Il1β. These results indicated that the anti-inflammatory effects of acute caffeine administration in the hippocampus may be mediated by a complex network of interdependencies between the Adora2B and Adora2A genes.
Collapse
Affiliation(s)
| | | | | | - Kassia de Carvalho
- Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
| | - Rogério Aires
- Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
| | - Jennyffer Souza
- Laboratory of Bioenergetics and Oxidative Stress - LABOX, Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Marcel Rodrigues Ferreira
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unity, Botucatu Medical School, São Paulo State University, Brazil.
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI, USA
| | - Maria Martha Bernardi
- Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
| | - Alexandra Latini
- Laboratory of Bioenergetics and Oxidative Stress - LABOX, Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Rodrigo A Foganholi da Silva
- Dentistry, University of Taubaté, Taubaté, São Paulo, São Paulo, Brazil; Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil.
| |
Collapse
|
4
|
Lopes CR, Gonçalves FQ, Olaio S, Tomé AR, Cunha RA, Lopes JP. Adenosine A 2A Receptors Shut Down Adenosine A 1 Receptor-Mediated Presynaptic Inhibition to Promote Implementation of Hippocampal Long-Term Potentiation. Biomolecules 2023; 13:biom13040715. [PMID: 37189461 DOI: 10.3390/biom13040715] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Adenosine operates a modulation system fine-tuning the efficiency of synaptic transmission and plasticity through A1 and A2A receptors (A1R, A2AR), respectively. Supramaximal activation of A1R can block hippocampal synaptic transmission, and the tonic engagement of A1R-mediated inhibition is increased with increased frequency of nerve stimulation. This is compatible with an activity-dependent increase in extracellular adenosine in hippocampal excitatory synapses, which can reach levels sufficient to block synaptic transmission. We now report that A2AR activation decreases A1R-medated inhibition of synaptic transmission, with particular relevance during high-frequency-induced long-term potentiation (LTP). Thus, whereas the A1R antagonist DPCPX (50 nM) was devoid of effects on LTP magnitude, the addition of an A2AR antagonist SCH58261 (50 nM) allowed a facilitatory effect of DPCPX on LTP to be revealed. Additionally, the activation of A2AR with CGS21680 (30 nM) decreased the potency of the A1R agonist CPA (6-60 nM) to inhibit hippocampal synaptic transmission in a manner prevented by SCH58261. These observations show that A2AR play a key role in dampening A1R during high-frequency induction of hippocampal LTP. This provides a new framework for understanding how the powerful adenosine A1R-mediated inhibition of excitatory transmission can be controlled to allow the implementation of hippocampal LTP.
Collapse
Affiliation(s)
- Cátia R Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco Q Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Simão Olaio
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Angelo R Tomé
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-534 Coimbra, Portugal
| | - João Pedro Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
5
|
Launay A, Nebie O, Vijaya Shankara J, Lebouvier T, Buée L, Faivre E, Blum D. The role of adenosine A 2A receptors in Alzheimer's disease and tauopathies. Neuropharmacology 2023; 226:109379. [PMID: 36572177 DOI: 10.1016/j.neuropharm.2022.109379] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Adenosine signals through four distinct G protein-coupled receptors that are located at various synapses, cell types and brain areas. Through them, adenosine regulates neuromodulation, neuronal signaling, learning and cognition as well as the sleep-wake cycle, all strongly impacted in neurogenerative disorders, among which Alzheimer's Disease (AD). AD is a complex form of cognitive deficits characterized by two pathological hallmarks: extracellular deposits of aggregated β-amyloid peptides and intraneuronal fibrillar aggregates of hyper- and abnormally phosphorylated Tau proteins. Both lesions contribute to the early dysfunction and loss of synapses which are strongly associated to the development of cognitive decline in AD patients. The present review focuses on the pathophysiological impact of the A2ARs dysregulation observed in cognitive area from AD patients. We are reviewing not only evidence of the cellular changes in A2AR levels in pathological conditions but also describe what is currently known about their consequences in term of synaptic plasticity, neuro-glial miscommunication and memory abilities. We finally summarize the proof-of-concept studies that support A2AR as credible targets and the clinical interest to repurpose adenosine drugs for the treatment of AD and related disorders. This article is part of the Special Issue on "Purinergic Signaling: 50 years".
Collapse
Affiliation(s)
- Agathe Launay
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Ouada Nebie
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Jhenkruthi Vijaya Shankara
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Thibaud Lebouvier
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France; CHU Lille, Memory Clinic, Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Emilie Faivre
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - David Blum
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, F-59000, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France.
| |
Collapse
|
6
|
Huang SY, Su ZY, Han YY, Liu L, Shang YJ, Mai ZF, Zeng ZW, Li CH. Cordycepin improved the cognitive function through regulating adenosine A 2A receptors in MPTP induced Parkinson's disease mice model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154649. [PMID: 36634379 DOI: 10.1016/j.phymed.2023.154649] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Parkinson's disease (PD), the most common neurodegenerative disorder, primarily affects dopaminergic neurons in the substantia nigra (SN). In addition to severe motor dysfunction, PD patients appear apparent cognitive impairments in the late stage. Cognitive dysfunction is accompanied by synaptic transmission damage in the hippocampus. Cordycepin has been reported to alleviate cognitive impairments in neurodegenerative diseases. PURPOSE The study aimed to estimate the protection roles of cordycepin on cognitive dysfunction in PD model and explore the potential mechanisms. METHODS 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was used to establish the PD model in vivo and in vitro experiments. In the in vivo experiments, the C57BL / 6 mice were intraperitoneally injected with MPTP and intragastric administration with cordycepin. Open field test (OFT) was used to estimate the exercise ability. Spontaneous alternation behavioral (SAB) and morris water maze (MWM) tests were used to evaluate the learning and memory abilities. The hippocampal slices from C57BL / 6 and Kunming mice in the in vitro experiments were used to record field excitatory postsynaptic potential (fEPSP) by electrophysiological methods. Western blotting was used to examine the level of tyrosine hydroxylase (TH) in the in vivo experiments and the levels of adenosine A1 and A2A receptors (A1R and A2AR) in the in vitro experiments, respectively. The drugs of MPTP, cordycepin, DPCPX and SCH58261 were perfused through dissolving in artificial cerebrospinal fluid. RESULTS Cordycepin could significantly reduce the impairments on motor, exploration, spatial learning and memory induce by MPTP. MPTP reduced the amplitude of LTP in hippocampal CA1 area but cordycepin could improve LTP amplitudes. Cordycepin at dosage of 20 mg/kg also increased the TH level in SN. In the in vitro experiments, MPTP inhibited synaptic transmission in hippocampal Schaffer-CA1 pathway with a dose-dependent relationship, while cordycepin could reverse the inhibition of synaptic transmission. Furthermore, the roles of cordycepin on synaptic transmission could been attenuated in the presence of the antagonists of A1R and A2AR, DPCPX and SCH58261, respectively. Interestingly, the level of A2AR rather than A1R in hippocampus was significantly decreased in the cordycepin group as compared to the control. CONCLUSION The present study has showed that cordycepin could improve cognitive function in the PD model induced by MPTP through regulating the adenosine A2A receptors. These findings were helpful to provide a new strategy for the dementia caused by Parkinson's disease.
Collapse
Affiliation(s)
- Shu-Yi Huang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Zong-Ying Su
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yuan-Yuan Han
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Li Liu
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Ying-Jie Shang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Zi-Fan Mai
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Zhi-Wei Zeng
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Chu-Hua Li
- School of Life Science, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
7
|
Huffels CFM, van Dijk RE, Karst H, Meye FJ, Hol EM, Middeldorp J. Systemic Injection of Aged Blood Plasma in Adult C57BL/6 Mice Induces Neurophysiological Impairments in the Hippocampal CA1. J Alzheimers Dis 2022; 89:283-297. [PMID: 35871343 DOI: 10.3233/jad-220337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Aging is characterized by systemic alterations and forms an important risk factor for Alzheimer's disease. Recently, it has been indicated that blood-borne factors present in the systemic milieu contribute to the aging process. Exposing young mice to aged blood plasma results in impaired neurogenesis and synaptic plasticity in the dentate gyrus, as well as impaired cognition. Vice versa, treating aged mice with young blood plasma rescues impairments associated with aging. OBJECTIVE Whether blood-borne factors are sufficient to drive impairments outside the dentate gyrus, how they impact neurophysiology, and how the functional outcome compares to impairments found in mouse models for AD is still unclear. METHODS Here, we treated adult mice with blood plasma from aged mice and assessed neurophysiological parameters in the hippocampal CA1. RESULTS Mice treated with aged blood plasma show significantly impaired levels of long-term potentiation (LTP), similar to those present in APP/PS1 mice. These impaired levels of LTP in plasma-treated mice are associated with alterations in basic properties of glutamatergic transmission and the enhanced activity of voltage-gated Ca2 + channels. CONCLUSION Together, the data presented in this study show that blood-borne factors are sufficient to drive neurophysiological impairments in the hippocampal CA1.
Collapse
Affiliation(s)
- Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Roland E van Dijk
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,Department of Neurobiology & Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| |
Collapse
|
8
|
Schubert C, Schulz K, Träger S, Plath AL, Omriouate A, Rosenkranz SC, Morellini F, Friese MA, Hirnet D. Neuronal Adenosine A1 Receptor is Critical for Olfactory Function but Unable to Attenuate Olfactory Dysfunction in Neuroinflammation. Front Cell Neurosci 2022; 16:912030. [PMID: 35846561 PMCID: PMC9279574 DOI: 10.3389/fncel.2022.912030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Adenine nucleotides, such as adenosine triphosphate (ATP), adenosine diphosphate (ADP), as well as the nucleoside adenosine are important modulators of neuronal function by engaging P1 and P2 purinergic receptors. In mitral cells, signaling of the G protein-coupled P1 receptor adenosine 1 receptor (A1R) affects the olfactory sensory pathway by regulating high voltage-activated calcium channels and two-pore domain potassium (K2P) channels. The inflammation of the central nervous system (CNS) impairs the olfactory function and gives rise to large amounts of extracellular ATP and adenosine, which act as pro-inflammatory and anti-inflammatory mediators, respectively. However, it is unclear whether neuronal A1R in the olfactory bulb modulates the sensory function and how this is impacted by inflammation. Here, we show that signaling via neuronal A1R is important for the physiological olfactory function, while it cannot counteract inflammation-induced hyperexcitability and olfactory deficit. Using neuron-specific A1R-deficient mice in patch-clamp recordings, we found that adenosine modulates spontaneous dendro-dendritic signaling in mitral and granule cells via A1R. Furthermore, neuronal A1R deficiency resulted in olfactory dysfunction in two separate olfactory tests. In mice with experimental autoimmune encephalomyelitis (EAE), we detected immune cell infiltration and microglia activation in the olfactory bulb as well as hyperexcitability of mitral cells and olfactory dysfunction. However, neuron-specific A1R activity was unable to attenuate glutamate excitotoxicity in the primary olfactory bulb neurons in vitro or EAE-induced olfactory dysfunction and disease severity in vivo. Together, we demonstrate that A1R modulates the dendro-dendritic inhibition (DDI) at the site of mitral and granule cells and impacts the processing of the olfactory sensory information, while A1R activity was unable to counteract inflammation-induced hyperexcitability.
Collapse
Affiliation(s)
- Charlotte Schubert
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Schulz
- Division of Neurophysiology, Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Simone Träger
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna-Lena Plath
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Asina Omriouate
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C. Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabio Morellini
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Manuel A. Friese,
| | - Daniela Hirnet
- Division of Neurophysiology, Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
- Daniela Hirnet,
| |
Collapse
|
9
|
Han MX, Jiang WY, Jiang Y, Wang LH, Xue R, Zhang GX, Chen JW. Gao-Zi-Yao improves learning and memory function in old spontaneous hypertensive rats. BMC Complement Med Ther 2022; 22:147. [PMID: 35643519 PMCID: PMC9148521 DOI: 10.1186/s12906-022-03630-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/18/2022] [Indexed: 11/21/2022] Open
Abstract
Aims Gao-Zi-Yao has long been a unique way for treating various diseases. The present study is to explore the effect of Gao-Zi-Yao on learning and memory function in old spontaneous hypertensive rats (SHR) and its possible mechanism. Method Male old SHR were received different doses of Gao-Zi-Yao for 4 weeks. Systolic blood pressure (SBP) and heart rate were monitored. Serum levels of nitric oxide (NO), interleukin (IL)-1β, IL-2, and tumor necrotic factor (TNF)-α were measured. Morris water maze was performed to test the learning and memory function of the rats. Number of neurons in hippocampus was counted by Nissl staining. Western blot was applied to detect the expressions of learning and memory function related proteins, N-methyl-d-aspartate receptor 2B (NMDAR 2B), glutamate receptor 1 (GluR1), phosphorylated-calmodulin-dependent protein kinase II (p-CaMK II), and phosphorylated-cAMP responsive element-binding protein (p-CREB) in rat hippocampus. Results Data showed that Gao-Zi-Yao reduced SBP in old SHR, elevated NO level, and suppressed levels of IL-1β, IL-2, TNF-α. The results of Morris water maze experiment showed that Gao-Zi-Yao dose-dependently improved learning and memory function. Number of neurons in the hippocampal dentate gyrus (DG) region of the old SHR was increased by Gao-Zi-Yao treatment. In addition, Gao-Zi-Yao elevated the protein expressions of NMDAR 2B, GluR1, p-CaMK II, and p-CREB in hippocampus. Conclusion Gao-Zi-Yao decreases SBP and improves the learning and memory function of the old SHR by regulation of oxidative stress, inflammatory factors and neuron number in hippocampal DG area and the expression of learning and memory function related proteins. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03630-0.
Collapse
|
10
|
Jakova E, Moutaoufik MT, Lee JS, Babu M, Cayabyab FS. Adenosine A1 receptor ligands bind to α-synuclein: implications for α-synuclein misfolding and α-synucleinopathy in Parkinson's disease. Transl Neurodegener 2022; 11:9. [PMID: 35139916 PMCID: PMC8830172 DOI: 10.1186/s40035-022-00284-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/21/2022] [Indexed: 12/20/2022] Open
Abstract
Background Accumulating α-synuclein (α-syn) aggregates in neurons and glial cells are the staples of many synucleinopathy disorders, such as Parkinson’s disease (PD). Since brain adenosine becomes greatly elevated in ageing brains and chronic adenosine A1 receptor (A1R) stimulation leads to neurodegeneration, we determined whether adenosine or A1R receptor ligands mimic the action of known compounds that promote α-syn aggregation (e.g., the amphetamine analogue 2-aminoindan) or inhibit α-syn aggregation (e.g., Rasagiline metabolite 1-aminoindan). In the present study, we determined whether adenosine, A1R receptor agonist N6-Cyclopentyladenosine (CPA) and antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) could directly interact with α-syn to modulate α-syn aggregation and neurodegeneration of dopaminergic neurons in the substantia nigra (SN). Methods Nanopore analysis and molecular docking were used to test the binding properties of CPA and DPCPX with α-syn in vitro. Sprague–Dawley rats were administered with 7-day intraperitoneal injections of the A1R ligands and 1- and 2-aminoindan, and levels of α-syn aggregation and neurodegeneration were examined in the SN pars compacta and hippocampal regions using confocal imaging and Western blotting. Results Using nanopore analysis, we showed that the A1R agonists (CPA and adenosine) interacted with the N-terminus of α-syn, similar to 2-aminoindan, which is expected to promote a “knot” conformation and α-syn misfolding. In contrast, the A1R antagonist DPCPX interacted with the N- and C-termini of α-syn, similar to 1-aminoindan, which is expected to promote a “loop” conformation that prevents α-syn misfolding. Molecular docking studies revealed that adenosine, CPA and 2-aminoindan interacted with the hydrophobic core of α-syn N-terminus, whereas DPCPX and 1-aminoindan showed direct binding to the N- and C-terminal hydrophobic pockets. Confocal imaging and Western blot analyses revealed that chronic treatments with CPA alone or in combination with 2-aminoindan increased α-syn expression/aggregation and neurodegeneration in both SN pars compacta and hippocampus. In contrast, DPCPX and 1-aminoindan attenuated the CPA-induced α-syn expression/aggregation and neurodegeneration in SN and hippocampus. Conclusions The results indicate that A1R agonists and drugs promoting a “knot” conformation of α-syn can cause α-synucleinopathy and increase neuronal degeneration, whereas A1R antagonists and drugs promoting a “loop” conformation of α-syn can be harnessed for possible neuroprotective therapies to decrease α-synucleinopathy in PD. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00284-3.
Collapse
Affiliation(s)
- Elisabet Jakova
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mohamed Taha Moutaoufik
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, SK, Canada
| | - Jeremy S Lee
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mohan Babu
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, SK, Canada
| | - Francisco S Cayabyab
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
11
|
Zarrinmayeh H, Territo PR. Purinergic Receptors of the Central Nervous System: Biology, PET Ligands, and Their Applications. Mol Imaging 2021; 19:1536012120927609. [PMID: 32539522 PMCID: PMC7297484 DOI: 10.1177/1536012120927609] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purinergic receptors play important roles in central nervous system (CNS). These receptors are involved in cellular neuroinflammatory responses that regulate functions of neurons, microglial and astrocytes. Based on their endogenous ligands, purinergic receptors are classified into P1 or adenosine, P2X and P2Y receptors. During brain injury or under pathological conditions, rapid diffusion of extracellular adenosine triphosphate (ATP) or uridine triphosphate (UTP) from the damaged cells, promote microglial activation that result in the changes in expression of several of these receptors in the brain. Imaging of the purinergic receptors with selective Positron Emission Tomography (PET) radioligands has advanced our understanding of the functional roles of some of these receptors in healthy and diseased brains. In this review, we have accumulated a list of currently available PET radioligands of the purinergic receptors that are used to elucidate the receptor functions and participations in CNS disorders. We have also reviewed receptors lacking radiotracer, laying the foundation for future discoveries of novel PET radioligands to reveal these receptors roles in CNS disorders.
Collapse
Affiliation(s)
- Hamideh Zarrinmayeh
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul R Territo
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
12
|
Mai ZF, Cao ZP, Huang SY, Yan WW, Huang JN, Wu BY, Li CH. The metaplastic effects of cordycepin in hippocampal CA1 area of rats. Eur J Pharmacol 2021; 897:173946. [PMID: 33607106 DOI: 10.1016/j.ejphar.2021.173946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/28/2021] [Accepted: 02/11/2021] [Indexed: 11/24/2022]
Abstract
Metaplasticity is referred to adjustment in the requirements for induction of synaptic plasticity based on the prior history of activity. Synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD), has been considered to be the neural processes underlying learning and memory. Previous observations that cordycepin (an adenosine derivative) improved learning and memory seemed to be contradictory to the findings that cordycepin inhibited LTP. Therefore, we speculated that the conflicting reports of cordycepin might be related to metaplasticity. In the current study, population spike (PS) in hippocampal CA1 area of rats was recorded by using electrophysiological method in vivo. The results showed that cordycepin reduced PS amplitude in hippocampal CA1 with a concentration-dependent relationship, and high frequency stimulation (HFS) failed to induce LTP when cordycepin was intrahippocampally administrated but improved LTP magnitude when cordycepin was pre-treated. Cordycepin increased LTD induced by activating N-Methyl-D-aspartate (NMDA) receptors and subsequently facilitated LTP induced by HFS. Furthermore, we found that 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), an adenosine A1 receptors antagonist, could block the roles of cordycepin on LTD and LTP. Collectively, cordycepin was able to modulate metaplasticity in hippocampal CA1 area of rats through adenosine A1 receptors. These findings would be helpful to reconcile the conflicting reports in the literatures and provided new insights into the mechanisms underlying cognitive function promotions with cordycepin treatment.
Collapse
Affiliation(s)
- Zi-Fan Mai
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Zhi-Ping Cao
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Shu-Yi Huang
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Wen-Wen Yan
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Jun-Ni Huang
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Bao-Yan Wu
- MOE Key Laboratory of Laser Life Science, Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| | - Chu-Hua Li
- School of Life Science, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
13
|
Adenosine Signaling and Clathrin-Mediated Endocytosis of Glutamate AMPA Receptors in Delayed Hypoxic Injury in Rat Hippocampus: Role of Casein Kinase 2. Mol Neurobiol 2021; 58:1932-1951. [PMID: 33415682 PMCID: PMC8018935 DOI: 10.1007/s12035-020-02246-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/03/2020] [Indexed: 11/20/2022]
Abstract
Chronic adenosine A1R stimulation in hypoxia leads to persistent hippocampal synaptic depression, while unopposed adenosine A2AR receptor stimulation during hypoxia/reperfusion triggers adenosine-induced post-hypoxia synaptic potentiation (APSP) and increased neuronal death. Still, the mechanisms responsible for this adenosine-mediated neuronal damage following hypoxia need to be fully elucidated. We tested the hypothesis that A1R and A2AR regulation by protein kinase casein kinase 2 (CK2) and clathrin-dependent endocytosis of AMPARs both contribute to APSPs and neuronal damage. The APSPs following a 20-min hypoxia recorded from CA1 layer of rat hippocampal slices were abolished by A1R and A2AR antagonists and by broad-spectrum AMPAR antagonists. The inhibitor of GluA2 clathrin-mediated endocytosis Tat-GluA2-3Y peptide and the dynamin-dependent endocytosis inhibitor dynasore both significantly inhibited APSPs. The CK2 antagonist DRB also inhibited APSPs and, like hypoxic treatment, caused opposite regulation of A1R and A2AR surface expression. APSPs were abolished when calcium-permeable AMPAR (CP-AMPAR) antagonist (IEM or philanthotoxin) or non-competitive AMPAR antagonist perampanel was applied 5 min after hypoxia. In contrast, perampanel, but not CP-AMPAR antagonists, abolished APSPs when applied during hypoxia/reperfusion. To test for neuronal viability after hypoxia, propidium iodide staining revealed significant neuroprotection of hippocampal CA1 pyramidal neurons when pretreated with Tat-GluA2-3Y peptide, CK2 inhibitors, dynamin inhibitor, CP-AMPAR antagonists (applied 5 min after hypoxia), and perampanel (either at 5 min hypoxia onset or during APSP). These results suggest that the A1R-CK2-A2AR signaling pathway in hypoxia/reperfusion injury model mediates increased hippocampal synaptic transmission and neuronal damage via calcium-permeable AMPARs that can be targeted by perampanel for neuroprotective stroke therapy.
Collapse
|
14
|
Jankowska A, Wesołowska A, Pawłowski M, Chłoń-Rzepa G. Multifunctional Ligands Targeting Phosphodiesterase as the Future Strategy for the Symptomatic and Disease-Modifying Treatment of Alzheimer’s Disease. Curr Med Chem 2020; 27:5351-5373. [DOI: 10.2174/0929867326666190620095623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023]
Abstract
Alzheimer’s Disease (AD) is a chronic neurodegenerative disorder characterized by cognitive
impairments such as memory loss, decline in language skills, and disorientation that affects
over 46 million people worldwide. Patients with AD also suffer from behavioral and psychological
symptoms of dementia that deteriorate their quality of life and lead to premature death. Currently
available drugs provide modest symptomatic relief but do not reduce pathological hallmarks (senile
plaques and neurofibrillary tangles) and neuroinflammation, both of which are integral parts of dementia.
A large body of evidence indicates that impaired signaling pathways of cyclic-3′,5′-
Adenosine Monophosphate (cAMP) and cyclic-3′,5′-guanosine Monophosphate (cGMP) may contribute
to the development and progression of AD. In addition, Phosphodiesterase (PDE) inhibitors,
commonly known as cAMP and/or cGMP modulators, were found to be involved in the phosphorylation
of tau; aggregation of amyloid beta; neuroinflammation; and regulation of cognition, mood,
and emotion processing. The purpose of this review was to update the most recent reports on the
development of novel multifunctional ligands targeting PDE as potential drugs for both symptomatic
and disease-modifying therapy of AD. This review collected the chemical structures of representative
multifunctional ligands, results of experimental in vitro and in vivo pharmacological studies,
and current opinions regarding the potential utility of these compounds for the comprehensive
therapy of AD. Finally, the multiparameter predictions of drugability of the representative compounds
were calculated and discussed.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Maciej Pawłowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| |
Collapse
|
15
|
Lv YC, Gao AB, Yang J, Zhong LY, Jia B, Ouyang SH, Gui L, Peng TH, Sun S, Cayabyab FS. Long-term adenosine A1 receptor activation-induced sortilin expression promotes α-synuclein upregulation in dopaminergic neurons. Neural Regen Res 2020; 15:712-723. [PMID: 31638096 PMCID: PMC6975149 DOI: 10.4103/1673-5374.266916] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Prolonged activation of adenosine A1 receptor likely leads to damage of dopaminergic neurons and subsequent development of neurodegenerative diseases. However, the pathogenesis underlying long-term adenosine A1 receptor activation-induced neurodegeneration remains unclear. In this study, rats were intraperitoneally injected with 5 mg/kg of the adenosine A1 receptor agonist N6-cyclopentyladenosine (CPA) for five weeks. The mobility of rats was evaluated by forced swimming test, while their cognitive capabilities were evaluated by Y-maze test. Expression of sortilin, α-synuclein, p-JUN, and c-JUN proteins in the substantia nigra were detected by western blot analysis. In addition, immunofluorescence staining of sortilin and α-synuclein was performed to detect expression in the substantia nigra. The results showed that, compared with adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (5 mg/kg) + CPA co-treated rats, motor and memory abilities were reduced, surface expression of sortin and α-synuclein in dopaminergic neurons was reduced, and total sortilin and total α-synuclein were increased in CPA-treated rats. MN9D cells were incubated with 500 nM CPA alone or in combination with 10 μM SP600125 (JNK inhibitor) for 48 hours. Quantitative real-time polymerase chain reaction analysis of sortilin and α-synuclein mRNA levels in MN9D cells revealed upregulated sortilin expression in MN9D cells cultured with CPA alone, but the combination of CPA and SP600125 could inhibit this expression. Predictions made using Jasper, PROMO, and Alibaba online databases identified a highly conserved sequence in the sortilin promoter that was predicted to bind JUN in both humans and rodents. A luciferase reporter assay of sortilin promoter plasmid-transfected HEK293T cells confirmed this prediction. After sortilin expression was inhibited by sh-SORT1, expression of p-JUN and c-JUN was detected by western blot analysis. Long-term adenosine A1 receptor activation levels upregulated α-synuclein expression at the post-transcriptional level by affecting sortilin expression. The online tool Raptor-X-Binding and Discovery Studio 4.5 prediction software predicted that sortilin can bind to α-synuclein. Co-immunoprecipitation revealed an interaction between sortilin and α-synuclein in MN9D cells. Our findings indicate that suppression of prolonged adenosine A1 receptor activation potently inhibited sortilin expression and α-synuclein accumulation, and dramatically improved host cognition and kineticism. This study was approved by the University Committee of Animal Care and Supply at the University of Saskatchewan (approval No. AUP#20070090) in March 2007 and the Animals Ethics Committee of University of South China (approval No. LL0387-USC) in June 2017.
Collapse
Affiliation(s)
- Yun-Cheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan Province, China; Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - An-Bo Gao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College; Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, Hunan Province, China
| | - Jing Yang
- Department of Metabolism & Endocrinology, the First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan Province, China
| | - Li-Yuan Zhong
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan Province, China
| | - Bo Jia
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan Province, China
| | - Shu-Hui Ouyang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan Province, China
| | - Le Gui
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Tian-Hong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan Province, China
| | - Sha Sun
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan Province, China
| | - Francisco S Cayabyab
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
16
|
Alves ACDB, Bristot VJDO, Limana MD, Speck AE, Barros LSD, Solano AF, Aguiar AS. Role of Adenosine A 2A Receptors in the Central Fatigue of Neurodegenerative Diseases. J Caffeine Adenosine Res 2019. [DOI: 10.1089/caff.2019.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Ana Cristina de Bem Alves
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | | | - Mirieli Denardi Limana
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | - Ana Elisa Speck
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | - Leonardo Soares de Barros
- LABOX—Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, UFSC—Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Alexandre Francisco Solano
- LABOX—Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, UFSC—Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Aderbal S. Aguiar
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| |
Collapse
|
17
|
Calker D, Biber K, Domschke K, Serchov T. The role of adenosine receptors in mood and anxiety disorders. J Neurochem 2019; 151:11-27. [DOI: 10.1111/jnc.14841] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Dietrich Calker
- Department for Psychiatry and Psychotherapy, Medical Center ‐ University of Freiburg, Faculty of Medicine University of Freiburg Freiburg Germany
| | - Knut Biber
- Section Medical Physiology, Department of Neuroscience University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Katharina Domschke
- Department for Psychiatry and Psychotherapy, Medical Center ‐ University of Freiburg, Faculty of Medicine University of Freiburg Freiburg Germany
- Centre for Basics in Neuromodulation, Faculty of Medicine University of Freiburg Freiburg Germany
| | - Tsvetan Serchov
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine, Medical Center ‐ University Freiburg University of Freiburg Freiburg Germany
| |
Collapse
|
18
|
Novel 8-amino-1,2,4-triazolo[4,3-a]pyrazin-3-one derivatives as potent human adenosine A 1 and A 2A receptor antagonists. Evaluation of their protective effect against β-amyloid-induced neurotoxicity in SH-SY5Y cells. Bioorg Chem 2019; 87:380-394. [PMID: 30913470 DOI: 10.1016/j.bioorg.2019.03.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/13/2018] [Accepted: 03/15/2019] [Indexed: 12/15/2022]
Abstract
In this work, an enlarged series of 1,2,4-triazolo[4,3-a]pyrazin-3-ones was designed to target the human (h) A2A adenosine receptor (AR) or both hA1 and hA2A ARs. The novel 8-amino-1,2,4-triazolopyrazin-3-one derivatives 1-25 featured a phenyl or a benzyl pendant at position 2 while different aryl/heteroaryl substituents were placed at position 6. Two compounds (8 and 10) endowed with high affinity (Ki = 7.2 and 10.6 nM) and a complete selectivity for the hA2A AR were identified. Moreover, several derivatives possessed nanomolar affinity for both hA1 and hA2A ARs (both Ki < 20 nM) and different degrees of selectivity versus the hA3 AR. Two selected compounds (10 and 25) demonstrated ability in preventing β-amyloid peptide (25-35)-induced neurotoxicity in SH-SY5Y cells. Results of docking studies at the hA2A and hA1 AR crystal structures helped us to rationalize the observed affinity data and to highlight that the steric hindrance of the substituents at the 2- and 6-position of the bicyclic core affects the binding mode in the receptor cavity.
Collapse
|
19
|
Frenguelli BG. The Purine Salvage Pathway and the Restoration of Cerebral ATP: Implications for Brain Slice Physiology and Brain Injury. Neurochem Res 2019; 44:661-675. [PMID: 28836168 PMCID: PMC6420432 DOI: 10.1007/s11064-017-2386-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 12/24/2022]
Abstract
Brain slices have been the workhorse for many neuroscience labs since the pioneering work of Henry McIlwain in the 1950s. Their utility is undisputed and their acceptance as appropriate models for the central nervous system is widespread, if not universal. However, the skeleton in the closet is that ATP levels in brain slices are lower than those found in vivo, which may have important implications for cellular physiology and plasticity. Far from this being a disadvantage, the ATP-impoverished slice can serve as a useful and experimentally-tractable surrogate for the injured brain, which experiences similar depletion of cellular ATP. We have shown that the restoration of cellular ATP in brain slices to in vivo values is possible with a simple combination of D-ribose and adenine (RibAde), two substrates for ATP synthesis. Restoration of ATP in slices to physiological levels has implications for synaptic transmission and plasticity, whilst in the injured brain in vivo RibAde shows encouraging positive results. Given that ribose, adenine, and a third compound, allopurinol, are all separately in use in man, their combined application after acute brain injury, in accelerating ATP synthesis and increasing the reservoir of the neuroprotective metabolite, adenosine, may help reduce the morbidity associated with stroke and traumatic brain injury.
Collapse
|
20
|
Cabrera-Pastor A, Taoro-González L, Cuñat AN, Canet-López D, Balzano T, Felipo V. Extracellular Cyclic GMP Modulates Membrane Expression of The GluA1 and GluA2 Subunits of AMPA Receptor in Cerebellum: Molecular Mechanisms Involved. Sci Rep 2017; 7:17656. [PMID: 29247190 PMCID: PMC5732250 DOI: 10.1038/s41598-017-18024-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/05/2017] [Indexed: 02/06/2023] Open
Abstract
There is increasing evidence that extracellular cGMP modulates glutamatergic neurotransmission and some forms of learning. However, the underlying mechanisms remain unknown. We proposed the hypotheses that extracellular cGMP may regulate membrane expression of AMPA receptors. To do this extracellular cGMP should act on a membrane protein and activate signal transduction pathways modulating phosphorylation of the GluA1 and/or GluA2 subunits. It has been shown that extracellular cGMP modulates glycine receptors. The aims of this work were to assess: 1) whether extracellular cGMP modulates membrane expression of GluA1 and GluA2 subunits of AMPA receptors in cerebellum in vivo; 2) whether this is mediated by glycine receptors; 3) the role of GluA1 and GluA2 phosphorylation and 4) identify steps of the intracellular pathways involved. We show that extracellular cGMP modulates membrane expression of GluA1 and GluA2 in cerebellum in vivo and unveil the mechanisms involved. Extracellular cGMP reduced glycine receptor activation, modulating cAMP, protein kinases and phosphatases, and GluA1 and GluA2 phosphorylation, resulting in increased GluA1 and reduced GluA2 membrane expression. Extracellular cGMP therefore modulates membrane expression of AMPA receptors and glutamatergic neurotransmission. The steps identified may be therapeutic targets to improve neurotransmission and neurological function in pathological situations with abnormal glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Andrea Cabrera-Pastor
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain.
| | - Lucas Taoro-González
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Amparo N Cuñat
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - David Canet-López
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Tiziano Balzano
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| |
Collapse
|
21
|
Bukhari SA, Saul MC, Seward CH, Zhang H, Bensky M, James N, Zhao SD, Chandrasekaran S, Stubbs L, Bell AM. Temporal dynamics of neurogenomic plasticity in response to social interactions in male threespined sticklebacks. PLoS Genet 2017; 13:e1006840. [PMID: 28704398 PMCID: PMC5509087 DOI: 10.1371/journal.pgen.1006840] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/27/2017] [Indexed: 11/18/2022] Open
Abstract
Animals exhibit dramatic immediate behavioral plasticity in response to social interactions, and brief social interactions can shape the future social landscape. However, the molecular mechanisms contributing to behavioral plasticity are unclear. Here, we show that the genome dynamically responds to social interactions with multiple waves of transcription associated with distinct molecular functions in the brain of male threespined sticklebacks, a species famous for its behavioral repertoire and evolution. Some biological functions (e.g., hormone activity) peaked soon after a brief territorial challenge and then declined, while others (e.g., immune response) peaked hours afterwards. We identify transcription factors that are predicted to coordinate waves of transcription associated with different components of behavioral plasticity. Next, using H3K27Ac as a marker of chromatin accessibility, we show that a brief territorial intrusion was sufficient to cause rapid and dramatic changes in the epigenome. Finally, we integrate the time course brain gene expression data with a transcriptional regulatory network, and link gene expression to changes in chromatin accessibility. This study reveals rapid and dramatic epigenomic plasticity in response to a brief, highly consequential social interaction. Social interactions provoke changes in the brain and behavior but their underlying molecular mechanisms remain obscure. Male sticklebacks are small fish whose fitness depends on their ability to defend a territory. Here, by measuring the time course of gene expression in response to a territorial challenge in two brain regions, we show that a single brief territorial intrusion provoked waves of gene expression that persisted for hours afterwards, with waves of transcription associated with distinct biological processes. Moreover, a single territorial challenge caused dramatic changes to the epigenome. Changes in chromatin accessibility corresponded to changes in gene expression, and to the activity of transcription factors operating within gene regulatory networks. This study reveals rapid and dramatic epigenomic plasticity in response to a brief, highly consequential social interaction. These results suggest that meaningful social interactions (even brief ones) can provoke waves of transcription and changes to the epigenome which lead to changes in neural functioning, and those changes are a mechanism by which animals update their assessment of their social world.
Collapse
Affiliation(s)
- Syed Abbas Bukhari
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
- Illinois Informatics Institute, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
| | - Michael C. Saul
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
| | - Christopher H. Seward
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
| | - Huimin Zhang
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
| | - Miles Bensky
- Program in Ecology, Evolution and Conservation Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
| | - Noelle James
- Neuroscience Program, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
| | - Sihai Dave Zhao
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
- Department of Statistics, University of Illinois, Urbana Champaign, Urbana, IL United States of America
| | - Sriram Chandrasekaran
- Harvard Society of Fellows, Harvard University, Cambridge, MA, United States of America
- Faculty of Arts and Sciences, Harvard University, Cambridge, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | - Lisa Stubbs
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
- Department of Cell and Developmental Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
| | - Alison M. Bell
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
- Program in Ecology, Evolution and Conservation Biology, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
- Neuroscience Program, University of Illinois, Urbana Champaign, Urbana, IL, United States of America
- * E-mail:
| |
Collapse
|
22
|
Adenosine A1 and A2A Receptors in the Brain: Current Research and Their Role in Neurodegeneration. Molecules 2017; 22:molecules22040676. [PMID: 28441750 PMCID: PMC6154612 DOI: 10.3390/molecules22040676] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022] Open
Abstract
The inhibitory adenosine A1 receptor (A1R) and excitatory A2A receptor (A2AR) are predominantly expressed in the brain. Whereas the A2AR has been implicated in normal aging and enhancing neurotoxicity in multiple neurodegenerative diseases, the inhibitory A1R has traditionally been ascribed to have a neuroprotective function in various brain insults. This review provides a summary of the emerging role of prolonged A1R signaling and its potential cross-talk with A2AR in the cellular basis for increased neurotoxicity in neurodegenerative disorders. This A1R signaling enhances A2AR-mediated neurodegeneration, and provides a platform for future development of neuroprotective agents in stroke, Parkinson’s disease and epilepsy.
Collapse
|
23
|
Kugathasan P, Waller J, Westrich L, Abdourahman A, Tamm JA, Pehrson AL, Dale E, Gulinello M, Sanchez C, Li Y. In vivo and in vitro effects of vortioxetine on molecules associated with neuroplasticity. J Psychopharmacol 2017; 31:365-376. [PMID: 27678087 DOI: 10.1177/0269881116667710] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neuroplasticity is fundamental for brain functions, abnormal changes of which are associated with mood disorders and cognitive impairment. Neuroplasticity can be affected by neuroactive medications and by aging. Vortioxetine, a multimodal antidepressant, has shown positive effects on cognitive functions in both pre-clinical and clinical studies. In rodent studies, vortioxetine increases glutamate neurotransmission, promotes dendritic branching and spine maturation, and elevates hippocampal expression of the activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) at the transcript level. The present study aims to assess the effects of vortioxetine on several neuroplasticity-related molecules in different experimental systems. Chronic (1 month) vortioxetine increased Arc/Arg3.1 protein levels in the cortical synaptosomes of young and middle-aged mice. In young mice, this was accompanied by an increase in actin-depolymerizing factor (ADF)/cofilin serine 3 phosphorylation without altering the total ADF/cofilin protein level, and an increase in the GluA1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor phosphorylation at serine 845 (S845) without altering serine 831 (S831) GluA1 phosphorylation nor the total GluA1 protein level. Similar effects were detected in cultured rat hippocampal neurons: Acute vortioxetine increased S845 GluA1 phosphorylation without changing S831 GluA1 phosphorylation or the total GluA1 protein level. These changes were accompanied by an increase in α subunit of Ca2+/calmodulin-dependent kinase (CaMKIIα) phosphorylation (at threonine 286) without changing the total CaMKIIα protein level in cultured neurons. In addition, chronic (1 month) vortioxetine, but not fluoxetine, restored the age-associated reduction in Arc/Arg3.1 and c-Fos transcripts in the frontal cortex of middle-aged mice. Taken together, these results demonstrated that vortioxetine modulates molecular targets that are related to neuroplasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yan Li
- 1 Lundbeck Research, Paramus, NJ, USA
| |
Collapse
|