1
|
Cardoso R, Cardoso FSDS, Ramalho BDS, Maria GDS, Cavalcanti RR, Taboada TB, de Almeida JS, Martinez AMB, de Almeida FM. Inosine Improves Functional Recovery and Cell Morphology Following Compressive Spinal Cord Injury in Mice. Neurotrauma Rep 2024; 5:957-968. [PMID: 39464528 PMCID: PMC11512092 DOI: 10.1089/neur.2024.0081] [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: 10/29/2024] Open
Abstract
Spinal cord injury (SCI) is one of the most serious conditions of the central nervous system, causing motor and sensory deficits that lead to a significant impairment in the quality of life. Previous studies have indicated that inosine can promote regeneration after SCI. Here we investigated the effects of inosine on the behavioral and morphological recovery after a compressive injury. Adult female C57BL/6 mice were subjected to laminectomy and spinal cord compression using a vascular clip. Inosine or saline injections were administered intraperitoneally, with the first dose performed 24 h after injury and daily for 7 days after injury. The mice were evaluated using Basso Mouse Scale (BMS), locomotor rating scale, and pinprick test for 8 weeks. At the end, the animals were anesthetized and euthanized, and the spinal cords were collected for morphological evaluation. Inosine-treated animals presented better results in the immunostaining for oligodendrocytes and in the number of myelinated fibers through semithin sections compared to saline-treated animals, showing that there was a greater preservation of the white matter. Analysis of the immunoreactivity of astrocytes and evaluation of the inflammatory profile with macrophage labeling revealed that the animals of the inosine group had a lower immunoreactivity when compared to control, which suggests a reduction of the glial scar and less inflammation, respectively, leading to a more favorable microenvironment for spinal cord regeneration. Indeed, inosine-treated animals scored higher on the BMS scale and presented better results on the pinprick test, indicating that the treatment contributed to motor and sensory recovery. After the animals were sacrificed, we obtained the electroneuromyography, where the inosine group showed a greater amplitude of the compound muscle action potential. These results indicate that inosine contributed to the regeneration process in the spinal cord of mice submitted to compressive injury and should be further investigated as a candidate for SCI therapy.
Collapse
Affiliation(s)
- Ricardo Cardoso
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Fellipe Soares dos Santos Cardoso
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Bruna dos Santos Ramalho
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Guilherme dos Santos Maria
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Roberta Ramos Cavalcanti
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Tiago Bastos Taboada
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Juliana Silva de Almeida
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Ana Maria Blanco Martinez
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
| | - Fernanda Martins de Almeida
- Laboratório de Neurodegeneração e Reparo—Departamento de Anatomia, Patológica—Hospital Universitário Clementino Fraga Filho/UFRJ, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas—ICB/UFRJ, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Du H, Li C, Gao R, Tan Y, Wang B, Peng Y, Yang N, Ning Y, Li P, Zhao Y, Zhou Y. Inhibition of the interaction between microglial adenosine 2A receptor and NLRP3 inflammasome attenuates neuroinflammation posttraumatic brain injury. CNS Neurosci Ther 2024; 30:e14408. [PMID: 37564004 PMCID: PMC10805470 DOI: 10.1111/cns.14408] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
AIMS Adenosine 2A receptor (A2A R) is widely expressed in the brain and plays important roles in neuroinflammation, and the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a crucial component of the innate immune system while the regulation of A2A R on it in the central nervous system (CNS) has not been clarified. METHODS The effects of microglial A2A R on NLRP3 inflammasome assembly and activation were investigated in wild-type, A2A R- or NLRP3-knockout primary microglia with pharmacological treatment. Microglial A2A R or NLRP3 conditional knockout mice were used to interrogate the effects of this regulation on neuroinflammation posttraumatic brain injury (TBI). RESULTS We found that A2A R directly interacted with NLRP3 and facilitated NLRP3 inflammasome assembly and activation in primary microglia while having no effects on mRNA levels of inflammasome components. Inhibition of the interaction via A2A R agonist or knockout attenuated inflammasome assembly and activation in vitro. In the TBI model, microglial A2A R and NLRP3 were co-expressed at high levels in microglia next to the peri-injured cortex, and abrogating of this interaction by microglial NLRP3 or A2A R conditional knockout attenuated the neurological deficits and neuropathology post-TBI via reducing the NLRP3 inflammasome activation. CONCLUSION Our results demonstrated that inhibition of the interaction between A2A R and NLRP3 in microglia could mitigate the NLRP3 inflammasome assembly and activation and ameliorate the neuroinflammation post-TBI. It provides new insights into the effects of A2A R on neuroinflammation regulation post-TBI and offers a potential target for the treatment of NLRP3 inflammasome-related CNS diseases.
Collapse
Affiliation(s)
- Hao Du
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- The General Hospital of Tibet Military CommandTibetChina
| | - Chang‐Hong Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Ruo‐Bing Gao
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Yan Tan
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Department of Pathophysiology, College of High‐Altitude Military MedicineArmy Medical UniversityChongqingChina
| | - Bo Wang
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Yan Peng
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Nan Yang
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
| | - Ya‐Lei Ning
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| | - Ping Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| | - Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| | - Yuan‐Guo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery and Daping HospitalArmy Medical UniversityChongqingChina
- Institute of Brain and IntelligenceArmy Medical UniversityChongqingChina
| |
Collapse
|
3
|
Bykowski EA, Petersson JN, Dukelow S, Ho C, Debert CT, Montina T, Metz GAS. Identification of Serum Metabolites as Prognostic Biomarkers Following Spinal Cord Injury: A Pilot Study. Metabolites 2023; 13:metabo13050605. [PMID: 37233646 DOI: 10.3390/metabo13050605] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
The assessment, management, and prognostication of spinal cord injury (SCI) mainly rely upon observer-based ordinal scales measures. 1H nuclear magnetic resonance (NMR) spectroscopy provides an effective approach for the discovery of objective biomarkers from biofluids. These biomarkers have the potential to aid in understanding recovery following SCI. This proof-of-principle study determined: (a) If temporal changes in blood metabolites reflect the extent of recovery following SCI; (b) whether changes in blood-derived metabolites serve as prognostic indicators of patient outcomes based on the spinal cord independence measure (SCIM); and (c) whether metabolic pathways involved in recovery processes may provide insights into mechanisms that mediate neural damage and repair. Morning blood samples were collected from male complete and incomplete SCI patients (n = 7) following injury and at 6 months post-injury. Multivariate analyses were used to identify changes in serum metabolic profiles and were correlated to clinical outcomes. Specifically, acetyl phosphate, 1,3,7-trimethyluric acid, 1,9-dimethyluric acid, and acetic acid significantly related to SCIM scores. These preliminary findings suggest that specific metabolites may serve as proxy measures of the SCI phenotype and prognostic markers of recovery. Thus, serum metabolite analysis combined with machine learning holds promise in understanding the physiology of SCI and aiding in prognosticating outcomes following injury.
Collapse
Affiliation(s)
- Elani A Bykowski
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Jamie N Petersson
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Sean Dukelow
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Chester Ho
- Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Chantel T Debert
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Tony Montina
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
4
|
Li C, Xiong W, Wan B, Kong G, Wang S, Wang Y, Fan J. Role of peripheral immune cells in spinal cord injury. Cell Mol Life Sci 2023; 80:2. [PMID: 36478290 PMCID: PMC9729325 DOI: 10.1007/s00018-022-04644-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/01/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
Secondary spinal cord injury is caused by an inflammatory response cascade, and the process is irreversible. The immune system, as a mediator of inflammation, plays an important role in spinal cord injury. The spinal cord retains its immune privilege in a physiological state. Hence, elucidating the mechanisms by which peripheral immune cells are recruited to the lesion site and function after spinal cord injury is meaningful for the exploration of clinical therapeutic targets. In this review, we provide an overview of the multifaceted roles of peripheral immune cells in spinal cord injury.
Collapse
Affiliation(s)
- Cong Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wu Xiong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bowen Wan
- Department of Orthopaedics, Subei People's Hospital of Jiangsu, Clinical Medical College of Yangzhou University, Yangzhou, China
| | - Guang Kong
- Nanjing Medical University, Nanjing, 210029, China
| | - Siming Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yingying Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
5
|
Seven YB, Allen LL, Ciesla MC, Smith KN, Zwick A, Simon AK, Holland AE, Santiago JV, Stefan K, Ross A, Gonzalez-Rothi EJ, Mitchell GS. Intermittent Hypoxia Differentially Regulates Adenosine Receptors in Phrenic Motor Neurons with Spinal Cord Injury. Neuroscience 2022; 506:38-50. [PMID: 36273657 DOI: 10.1016/j.neuroscience.2022.10.007] [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: 06/28/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
Abstract
Cervical spinal cord injury (cSCI) impairs neural drive to the respiratory muscles, causing life- threatening complications such as respiratory insufficiency and diminished airway protection. Repetitive "low dose" acute intermittent hypoxia (AIH) is a promising strategy to restore motor function in people with chronic SCI. Conversely, "high dose" chronic intermittent hypoxia (CIH; ∼8 h/night), such as experienced during sleep apnea, causes pathology. Sleep apnea, spinal ischemia, hypoxia and neuroinflammation associated with cSCI increase extracellular adenosine concentrations and activate spinal adenosine receptors which in turn constrains the functional benefits of therapeutic AIH. Adenosine 1 and 2A receptors (A1, A2A) compete to determine net cAMP signaling and likely the tAIH efficacy with chronic cSCI. Since cSCI and intermittent hypoxia may regulate adenosine receptor expression in phrenic motor neurons, we tested the hypotheses that: 1) daily AIH (28 days) downregulates A2A and upregulates A1 receptor expression; 2) CIH (28 days) upregulates A2A and downregulates A1 receptor expression; and 3) cSCI alters the impact of CIH on adenosine receptor expression. Daily AIH had no effect on either adenosine receptor in intact or injured rats. However, CIH exerted complex effects depending on injury status. Whereas CIH increased A1 receptor expression in intact (not injured) rats, it increased A2A receptor expression in spinally injured (not intact) rats. The differential impact of CIH reinforces the concept that the injured spinal cord behaves in distinct ways from intact spinal cords, and that these differences should be considered in the design of experiments and/or new treatments for chronic cSCI.
Collapse
Affiliation(s)
- Yasin B Seven
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Latoya L Allen
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Marissa C Ciesla
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Kristin N Smith
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Amanda Zwick
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Alec K Simon
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashley E Holland
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Juliet V Santiago
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Kelsey Stefan
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashley Ross
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Elisa J Gonzalez-Rothi
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
6
|
Shalaby HN, Zaki HF, Ain-Shoka AAA, Mohammed RA. Adenosine A 2A Receptor Blockade Ameliorates Mania Like Symptoms in Rats: Signaling to PKC-α and Akt/GSK-3β/β-Catenin. Mol Neurobiol 2022; 59:6397-6410. [PMID: 35943710 PMCID: PMC9463338 DOI: 10.1007/s12035-022-02977-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/26/2022] [Indexed: 12/02/2022]
Abstract
Adenosinergic system dysfunction is implicated in the pathophysiology of multiple neuropsychiatric disorders including mania and bipolar diseases. The established synergistic interaction between A2A and D2 receptors in the prefrontal cortex could highlight the idea of A2A receptor antagonism as a possible anti-manic strategy. Hence, the present study was performed to examine the effect of a selective adenosine A2A receptor blocker (SCH58261) on methylphenidate-induced mania-like behavior while investigating the underlying mechanisms. Rats were injected with methylphenidate (5 mg/kg/day, i.p.) for 3 weeks with or without administration of either SCH58261 (0.01 mg/kg/day, i.p.) or lithium (150 mg/kg/day, i.p.) starting from day 9. In the diseased rats, adenosine A2AR antagonism reduced locomotor hyperactivity and risk-taking behavior along with decreased dopamine and glutamate levels. Meanwhile, SCH58261 restored NMDA receptor function, suppressed PKC-α expression, down-regulated β-Arrestin-2, up-regulated pS473-Akt and pS9-GSK-3β. Further, SCH58261 promoted synaptic plasticity markers through increasing BDNF levels along with down-regulating GAP-43 and SNAP-25. The A2A antagonist also reduced NF-κBp65 and TNF-α together with elevating IL-27 level giving an anti-inflammatory effect. In conclusion, suppression of PKC-α and modulation of Akt/GSK-3β/β-catenin axis through A2AR inhibition, could introduce adenosine A2AR as a possible therapeutic target for treatment of mania-like behavior. This notion is supported by the ability of the A2AR antagonist (SCH58261) to produce comparable results to those observed with the standard anti-manic drug (Lithium).
Collapse
Affiliation(s)
- Heba Nasr Shalaby
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Hala Fahmy Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | | | - Reham Atef Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
7
|
Corrigendum: Purinergic signaling systems across comparative models of spinal cord injury. Neural Regen Res 2022; 18:689-696. [PMID: 36018196 PMCID: PMC9727416 DOI: 10.4103/1673-5374.350234] [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] [Indexed: 12/15/2022] Open
Abstract
[This corrects the article DOI: 10.4103/1673-5374.338993].
Collapse
|
8
|
Soliño M, Larrayoz IM, López EM, Rey-Funes M, Bareiro M, Loidl CF, Girardi E, Martínez A, López-Costa JJ. Adenosine A2A Receptor: A New Neuroprotective Target in Light-Induced Retinal Degeneration. Front Pharmacol 2022; 13:840134. [PMID: 35387355 PMCID: PMC8977837 DOI: 10.3389/fphar.2022.840134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 11/24/2022] Open
Abstract
Continuous illumination induces the degeneration of photoreceptors. This animal model of light-induced retinal degeneration resembles many characteristics of human degenerative diseases of the outer retina, such as age-related macular degeneration. This work aimed to evaluate the potential neuroprotective effect of the modulation of adenosine A2A receptor in the model of light-induced retinal degeneration. Sprague-Dawley rats were intravitreally injected in the right eye with either CGS 21680, an adenosine A2A receptor agonist, or SCH 58261, an adenosine A2A receptor antagonist. Contralateral eyes were injected with respective vehicles as control. Then, rats were subjected to continuous illumination (12,000 lux) for 24 h. Retinas were processed by glial fibrillary acidic protein (GFAP) immunohistochemistry, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) technique, Western blotting (WB), and quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Another group of rats was subjected to functional studies by electroretinography. Animals treated with CGS21680 showed a significant increase of apoptotic nuclei in the outer nuclear layer and a significant increase of GFAP immunoreactive area of the retinas but did not alter WB nor electroretinography results. qRT-PCR showed that CGS 21680 significantly increased the expression of interleukin-1β. On the opposite, SCH 58261 significantly decreased apoptotic nuclei in the outer nuclear layer and GFAP immunoreactive area of the retinas. It also significantly decreased GFAP and activated caspase-3 levels as measured by WB and preserved retinal function, as treated eyes showed significantly greater amplitudes of a- and b-waves and oscillatory potentials. qRT-PCR revealed that SCH 58261 significantly decreased the expression of tumor necrosis factor-α. These results show that the blockade of the A2A receptor before the start of the pathogenic process is neuroprotective, as it prevents light-induced retinal damage. The use of A2A receptor antagonists deserves to be evaluated in retinal degenerative diseases.
Collapse
Affiliation(s)
- Manuel Soliño
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Ester María López
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Manuel Rey-Funes
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Mariana Bareiro
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Cesar Fabián Loidl
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Elena Girardi
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Alfredo Martínez
- Angiogenesis Study Group, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Juan José López-Costa
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
9
|
Thuraiaiyah J, Kokoti L, Al-Karagholi MAM, Ashina M. Involvement of adenosine signaling pathway in migraine pathophysiology: a systematic review of preclinical studies. J Headache Pain 2022; 23:43. [PMID: 35382738 PMCID: PMC8981838 DOI: 10.1186/s10194-022-01412-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/10/2022] [Indexed: 12/14/2022] Open
Abstract
Background Adenosine is a purinergic signaling molecule with a wide range of physiological functions including anti- and pronociceptive properties. Adenosine receptors are expressed in the trigeminovascular system, and adenosine receptor antagonist, caffeine, relieves migraine headache. We performed a systematic review of the literature of preclinical data addressing the role of adenosine in migraine pathophysiology. Methods PubMed and EMBASE were searched for pre-clinical studies on the role of adenosine in migraine pathophysiology on September 5th, 2021. Results A total of 2510 studies were screened by title and abstract. Of these, thirteen pre-clinical studies evaluating adenosine, adenosine A1, A2A and A3 receptors were included. These studies showed that adenosine signaling pathway is involved in controlling vascular tone. Furthermore, electrical stimulation of the trigeminal ganglion modulates the expression of adenosine A1 and A2A receptors in the trigeminal ganglion and trigeminal nucleus caudalis implicating adenosine signaling pathway in pain transmission. Conclusion Preclinical studies showed that adenosine has a dual effect on vasodilation and trigeminal pain pathway due to different receptor activation, suggesting a possible role of adenosine in migraine pathophysiology. Studies investigating pharmacological characteristics of subtypes of adenosine receptors are needed to further elucidate their role as a potential target for migraine treatment.
Collapse
|
10
|
Thuraiaiyah J, Kokoti L, Al-Karagholi MAM, Ashina M. Involvement of adenosine signaling pathway in migraine pathophysiology: A systematic review of clinical studies. Cephalalgia 2022; 42:781-792. [PMID: 35301855 DOI: 10.1177/03331024221077665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To systematically review clinical studies investigating the involvement of adenosine and its receptors in migraine pathophysiology. BACKGROUND Adenosine is a purinergic signaling molecule, clinically used in cardiac imaging during stress tests. Headache is a frequent adverse event after intravenous adenosine administration. Migraine headache relief is reported after intake of adenosine receptor antagonist, caffeine. These findings suggest a possible involvement of adenosine signaling in migraine pathophysiology and its potential as a drug target. METHODS A search through PubMed and EMBASE was undertaken for clinical studies investigating the role of adenosine and its receptors in migraine, published until September 2021. RESULTS A total of 2510 studies were screened by title and abstract. Of these, seven clinical studies were included. The main findings were that adenosine infusion induced headache, and plasma adenosine levels were elevated during ictal compared to interictal periods in migraine patients. CONCLUSION The present systematic review emphasizes a potentially important role of adenosine signaling in migraine pathogenesis. Further randomized and placebo-controlled clinical investigations applying adenosine receptors modulators in migraine patients are needed to further understand the adenosine involvement in migraine.
Collapse
Affiliation(s)
- Janu Thuraiaiyah
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lili Kokoti
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Mohammad Al-Mahdi Al-Karagholi
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Danish Headache Knowledge Center, Rigshospitalet - Glostrup, Glostrup, Denmark
| |
Collapse
|
11
|
Mou KJ, Shen KF, Li YL, Wu ZF, Duan W. Adenosine A 2A Receptor in Bone Marrow-Derived Cells Mediated Macrophages M2 Polarization via PPARγ-P65 Pathway in Chronic Hypoperfusion Situation. Front Aging Neurosci 2022; 13:792733. [PMID: 35046793 PMCID: PMC8761901 DOI: 10.3389/fnagi.2021.792733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The role of adenosine A2A receptor (A2AR) in the ischemic white matter damage induced by chronic cerebral hypoperfusion remains obscure. Here we investigated the role of A2AR in the process of macrophage polarizations in the white matter damage induced by chronic cerebral hypoperfusion and explored the involved signaling pathways. Methods: We combined mouse model and macrophage cell line for our study. White matter lesions were induced in A2AR knockout mice, wild-type mice, and chimeric mice generated by bone marrow cells transplantation through bilateral common carotid artery stenosis. Microglial/macrophage polarization in the corpus callosum was detected by immunofluorescence. For the cell line experiments, RAW264.7 macrophages were treated with the A2AR agonist CHS21680 or A2AR antagonist SCH58261 for 30 min and cultured under low-glucose and hypoxic conditions. Macrophage polarization was examined by immunofluorescence. The expression of peroxisome proliferator activated receptor gamma (PPARγ) and transcription factor P65 was examined by western blotting and real-time polymerase chain reaction (RT-PCR). Inflammatory cytokine factors were assessed by enzyme-linked immunosorbent assay (ELISA) and RT-PCR. Results: Both global A2AR knockout and inactivation of A2AR in bone marrow-derived cells enhanced M1 marker expression in chronic ischemic white matter lesions. Under low-glucose and hypoxic conditions, CGS21680 treatment promoted macrophage M2 polarization, increased the expression of PPARγ, P65, and interleukin-10 (IL-10) and suppressed the expression of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). The CGS21680-induced upregulation of P65 and IL-10 was abolished in macrophages upon PPARγ knockdown. The downregulation of TNF-α and IL-1β by CGS21680 was less affected by PPARγ knockdown. Conclusions: In the cerebral hypoperfusion induced white matter damage, A2AR signaling in bone marrow-derived cells induces macrophage M2 polarization and increases the expression of the anti-inflammatory factor IL-10 via the PPARγ-P65 pathway, both of which might explain its neuroprotective effect.
Collapse
Affiliation(s)
- Ke-Jie Mou
- Department of Neurosurgery, Bishan Hospital of Chongqing, Chongqing, China
| | - Kai-Feng Shen
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yan-Ling Li
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zhi-Feng Wu
- Department of Pediatrics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wei Duan
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Chen L, Zhou Y, Wang J, Li K, Zhang Z, Peng M. The adenosine A 2A receptor alleviates postoperative delirium-like behaviors by restoring blood cerebrospinal barrier permeability in rats. J Neurochem 2021; 158:980-996. [PMID: 34033116 DOI: 10.1111/jnc.15436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
Postoperative delirium (POD) is a common post-operative complication in elderly patients that is associated with increased morbidity and mortality. However, the neuropathogenesis of this complication remains unknown. The blood-cerebrospinal fluid barrier (BCB) and brain-blood barrier (BBB) are composed of tight junctions between cells that form physical barriers, and BBB damage plays an important role in the neuropathogenesis of POD. Nevertheless, the role of BCB in POD remains to be elucidated. Herein, we investigated the effect of adenosine A2A receptor (A2A R), a key regulator of the permeability of barriers, on surgery-induced increased permeability of BCB and POD-like behaviors. Open field, buried food, and Y maze tests were used to evaluate behavioral changes in rats after surgery. Levels of tight junction proteins, adherens junction proteins, A2A R, GTP-RhoA, and ROCK2 in the choroid plexus were assessed by western blotting. The concentrations of NaFI and FITC-dextran in the cerebrospinal fluid (CSF) were detected by fluorescence spectrophotometry. Transmission electron microscopy was applied to observe the ultrastructure of the choroid plexus. Surgery/anesthesia decreased the levels of tight junction (e.g., ZO-1, occludin, and claudin1) proteins, increased concentrations of NaFI and FITC-dextran in CSF, damaged the ultrastructure of choroid plexus, and induced POD-like behaviors in rats. An A2A R antagonist alleviated POD-like behaviors in rats. Furthermore, the A2A R antagonist increased the levels of tight junction proteins and restored the permeability of BCB in rats with POD. Fasudil, a selective Rho-associated protein kinase 2 (ROCK2) inhibitor, ameliorated POD-like behaviors induced by A2A R activation. Moreover, fasudil also abolished the increased levels of GTP-RhoA/ROCK2, decreased levels of tight junction proteins, and increased permeability of BCB caused by A2A R activation. Our findings demonstrate that A2A R might participate in regulating BCB permeability in rats with POD via the RhoA/ROCK2 signaling pathway, which suggests the potential of A2A R as a therapeutic target for POD.
Collapse
Affiliation(s)
- Lei Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiayu Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ke Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mian Peng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Fakhri S, Abbaszadeh F, Jorjani M. On the therapeutic targets and pharmacological treatments for pain relief following spinal cord injury: A mechanistic review. Biomed Pharmacother 2021; 139:111563. [PMID: 33873146 DOI: 10.1016/j.biopha.2021.111563] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is globally considered as one of the most debilitating disorders, which interferes with daily activities and life of the affected patients. Despite many developments in related recognizing and treating procedures, post-SCI neuropathic pain (NP) is still a clinical challenge for clinicians with no distinct treatments. Accordingly, a comprehensive search was conducted in PubMed, Medline, Scopus, Web of Science, and national database (SID and Irandoc). The relevant articles regarding signaling pathways, therapeutic targets and pharmacotherapy of post-SCI pain were also reviewed. Data were collected with no time limitation until November 2020. The present study provides the findings on molecular mechanisms and therapeutic targets, as well as developing the critical signaling pathways to introduce novel neuroprotective treatments of post-SCI pain. From the pathophysiological mechanistic point of view, post-SCI inflammation activates the innate immune system, in which the immune cells elicit secondary injuries. So, targeting the critical signaling pathways for pain management in the SCI population has significant importance in providing new treatments. Indeed, several receptors, ion channels, excitatory neurotransmitters, enzymes, and key signaling pathways could be used as therapeutic targets, with a pivotal role of n-methyl-D-aspartate, gamma-aminobutyric acid, and inflammatory mediators. The current review focuses on conventional therapies, as well as crucial signaling pathways and promising therapeutic targets for post-SCI pain to provide new insights into the clinical treatment of post-SCI pain. The need to develop innovative delivery systems to treat SCI is also considered.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Abbaszadeh
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Masoumeh Jorjani
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Boknik P, Eskandar J, Hofmann B, Zimmermann N, Neumann J, Gergs U. Role of Cardiac A 2A Receptors Under Normal and Pathophysiological Conditions. Front Pharmacol 2021; 11:627838. [PMID: 33574762 PMCID: PMC7871008 DOI: 10.3389/fphar.2020.627838] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
This review presents an overview of cardiac A2A-adenosine receptors The localization of A2A-AR in the various cell types that encompass the heart and the role they play in force regulation in various mammalian species are depicted. The putative signal transduction systems of A2A-AR in cells in the living heart, as well as the known interactions of A2A-AR with membrane-bound receptors, will be addressed. The possible role that the receptors play in some relevant cardiac pathologies, such as persistent or transient ischemia, hypoxia, sepsis, hypertension, cardiac hypertrophy, and arrhythmias, will be reviewed. Moreover, the cardiac utility of A2A-AR as therapeutic targets for agonistic and antagonistic drugs will be discussed. Gaps in our knowledge about the cardiac function of A2A-AR and future research needs will be identified and formulated.
Collapse
Affiliation(s)
- P. Boknik
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Münster, Germany
| | - J. Eskandar
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Münster, Germany
| | - B. Hofmann
- Cardiac Surgery, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - N. Zimmermann
- Bundesinstitut für Arzneimittel und Medizinprodukte, Bonn, Germany
| | - J. Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - U. Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| |
Collapse
|
15
|
Zhao J, Kumar M, Sharma J, Yuan Z. Arbutin effectively ameliorates the symptoms of Parkinson's disease: the role of adenosine receptors and cyclic adenosine monophosphate. Neural Regen Res 2021; 16:2030-2040. [PMID: 33642391 PMCID: PMC8343309 DOI: 10.4103/1673-5374.308102] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
An antagonistic communication exists between adenosinergic and dopaminergic signaling in the basal ganglia, which suggests that the suppression of adenosine A2A receptors-cyclic adenosine monophosphate pathway may be able to restore the disrupted dopamine transmission that results in motor symptoms in Parkinson’s disease (PD). Arbutin is a natural glycoside that possesses antioxidant, anti-inflammatory, and neuroprotective properties. The purpose of this study was to investigate whether arbutin could ameliorate the symptoms of PD and to examine the underlying mechanism. In this study, Swiss albino mouse models of PD were established by the intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine for 4 successive days, with the concurrent intraperitoneal administration of arbutin (50 and 100 mg/kg) for 7 days. The results showed that arbutin significantly reduced lipid peroxidation, total nitrite levels, and inflammation in the substantia nigra and striatum of PD mouse models. In addition, arbutin decreased the activity of endogenous antioxidants, reduced the levels of dopamine, 3,4-dihydroxyphenylacetic acid, homovanillic acid, and γ-aminobutyric acid, and minimized neurodegeneration in the striatum. Arbutin also reduced the abnormal performance of PD mouse models in the open field test, bar test, pole test, and rotarod test. The therapeutic efficacy of arbutin was similar to that of madopar. The intraperitoneal injection of the A2AR agonist CGS21680 (0.5 mg/kg) attenuated the therapeutic effects of arbutin, whereas the intraperitoneal injection of forskolin (3 mg/kg) enhanced arbutin-mediated improvements. These findings suggest that arbutin can improve the performance of PD mouse models by inhibiting the function of the A2AR and enhancing the effects of cyclic adenosine monophosphate. This study was approved by the Institutional Animal Ethics Committee (1616/PO/Re/S/12/CPCSEA) on November 17, 2019 (approval No. IAEC/2019/010).
Collapse
Affiliation(s)
- Jie Zhao
- Department of Neurology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Manish Kumar
- Department of Pharmacology, Swift School of Pharmacy, Rajpura (Patiala), Punjab, India
| | - Jeevan Sharma
- Department of Pharmacology, Swift School of Pharmacy, Rajpura (Patiala), Punjab, India
| | - Zhihai Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
16
|
Dokalis N, Prinz M. Resolution of neuroinflammation: mechanisms and potential therapeutic option. Semin Immunopathol 2019; 41:699-709. [PMID: 31705317 DOI: 10.1007/s00281-019-00764-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/12/2019] [Indexed: 01/01/2023]
Abstract
The central nervous system (CNS) is comprised by an elaborate neural network that is under constant surveillance by tissue-intrinsic factors for maintenance of its homeostasis. Invading pathogens or sterile injuries might compromise vitally the CNS integrity and function. A prompt anti-inflammatory response is therefore essential to contain and repair the local tissue damage. Although the origin of the insults might be different, the principles of tissue backlashes, however, share striking similarities. CNS-resident cells, such as microglia and astrocytes, together with peripheral immune cells orchestrate an array of events that aim to functional restoration. If the acute inflammatory event remains unresolved, it becomes toxic leading to progressive CNS degeneration. Therefore, the cellular, molecular, and biochemical processes that regulate inflammation need to be on a fine balance with the intrinsic CNS repair mechanisms that influence tissue healing. The purpose of this review is to highlight aspects that facilitate the resolution of CNS inflammation, promote tissue repair, and functional recovery after acute injury and infection that could potentially contribute as therapeutic interventions.
Collapse
Affiliation(s)
- Nikolaos Dokalis
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany. .,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany. .,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
17
|
Seven YB, Simon AK, Sajjadi E, Zwick A, Satriotomo I, Mitchell GS. Adenosine 2A receptor inhibition protects phrenic motor neurons from cell death induced by protein synthesis inhibition. Exp Neurol 2019; 323:113067. [PMID: 31629857 DOI: 10.1016/j.expneurol.2019.113067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/12/2019] [Accepted: 09/18/2019] [Indexed: 01/31/2023]
Abstract
Respiratory motor neuron survival is critical for maintenance of adequate ventilation and airway clearance, preventing dependence to mechanical ventilation and respiratory tract infections. Phrenic motor neurons are highly vulnerable in rodent models of motor neuron disease versus accessory inspiratory motor pools (e.g. intercostals, scalenus). Thus, strategies that promote phrenic motor neuron survival when faced with disease and/or toxic insults are needed to help preserve breathing ability, airway defense and ventilator independence. Adenosine 2A receptors (A2A) are emerging as a potential target to promote neuroprotection, although their activation can have both beneficial and pathogenic effects. Since the role of A2A receptors in the phrenic motor neuron survival/death is not known, we tested the hypothesis that A2A receptor antagonism promotes phrenic motor neuron survival and preserves diaphragm function when faced with toxic, neurodegenerative insults that lead to phrenic motor neuron death. We utilized a novel neurotoxic model of respiratory motor neuron death recently developed in our laboratory: intrapleural injections of cholera toxin B subunit (CtB) conjugated to the ribosomal toxin, saporin (CtB-Saporin). We demonstrate that intrapleural CtB-Saporin causes: 1) profound phrenic motor neuron death (~5% survival); 2) ~7-fold increase in phrenic motor neuron A2A receptor expression prior to cell death; and 3) diaphragm muscle paralysis (inactive in most rats; ~7% residual diaphragm EMG amplitude during room air breathing). The A2A receptor antagonist istradefylline given after CtB-Saporin: 1) reduced phrenic motor neuron death (~20% survival) and 2) preserved diaphragm EMG activity (~46%). Thus, A2A receptors contribute to neurotoxic phrenic motor neuron death, an effect mitigated by A2A receptor antagonism.
Collapse
Affiliation(s)
- Yasin B Seven
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Alec K Simon
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Elaheh Sajjadi
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Amanda Zwick
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Irawan Satriotomo
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Gordon S Mitchell
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
18
|
Carbamazepine Attenuates Astroglial L-Glutamate Release Induced by Pro-Inflammatory Cytokines via Chronically Activation of Adenosine A 2A Receptor. Int J Mol Sci 2019; 20:ijms20153727. [PMID: 31366130 PMCID: PMC6695599 DOI: 10.3390/ijms20153727] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/18/2022] Open
Abstract
Carbamazepine (CBZ) binds adenosine receptors, but detailed effects of CBZ on astroglial transmission associated with adenosine receptor still need to be clarified. To clarify adenosinergic action of CBZ on astroglial transmission, primary cultured astrocytes were acutely or chronically treated with CBZ, proinflammatory cytokines (interferon γ (IFNγ) and tumor necrosis factor α (TNFα)), and adenosine A2A receptor (A2AR) agonist (CGS21680). IFNγ and TNFα increased basal, adenophostin-A (AdA)-evoked, and 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid (AMPA)-evoked astroglial L-glutamate releases. In physiological condition, CGS21680 increased basal astroglial L-glutamate release but glutamate transporter inhibition prevented this CGS21680 action. CBZ did not affect basal release, whereas glutamate transporter inhibition generated CBZ-induced glutamate release. Furthermore, AdA-evoked and AMPA-evoked releases were inhibited by CBZ but were unaffected by CGS21680. Contrary to physiological condition, chronic administrations of IFNγ and TNFα enhanced basal, AdA-, and AMPA-evoked releases, whereas IFNγ and TNFα decreased and increased CGS21680-evoked releases via modulation A2AR expression. Both chronic administration of CGS21680 and CBZ suppressed astroglial L-glutamate release responses induced by chronic cytokine exposer. Especifically, chronic administration of CBZ and CGS21680 prevented the reduction and elevation of A2AR expression by respective IFNγ and TNFα. These findings suggest that A2AR agonistic effects of CBZ contribute to chronic prevention of pathomechanisms developments of several neuropsychiatric disorders associated with proinflammatory cytokines.
Collapse
|
19
|
Liu Z, Zhang H, Xia H, Wang B, Zhang R, Zeng Q, Guo L, Shen K, Wang B, Zhong Y, Li Z, Sun G. CD8 T cell-derived perforin aggravates secondary spinal cord injury through destroying the blood-spinal cord barrier. Biochem Biophys Res Commun 2019; 512:367-372. [PMID: 30894275 DOI: 10.1016/j.bbrc.2019.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 03/01/2019] [Indexed: 12/27/2022]
Abstract
Perforin plays an important role in autoimmune and infectious diseases, but its function in immune inflammatory responses after spinal cord injury (SCI) has received insufficient attention. The goal of this study is to determine the influence of perforin after spinal cord injury (SCI) on secondary inflammation. Compared recovery from SCI in perforin knockout (Prf1-/-) and wild-type(WT)mice, WT mice had significantly lower the Basso mouse score (BMS), CatWalk XT, and motor-evoked potentials (MEPs) than Prf1-/- mice. Spinal cord lesions were also more obvious through glial fibrillary acidic protein (GFAP), Nissl, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining. Furthermore, the blood-spinal cord barrier (BSCB) disruption was more severe and inflammatory cytokine levels were higher. Flow cytometry indicated that perforin mainly originated from CD8 T cells. With flow cytometry and enzyme-linked immunosorbent assay (ELISA), human cerebrospinal fluid (CSF) yielded similar results. Together, this study firstly demonstrated that CD8 T cell-derived perforin is detrimental to SCI recovery in the mouse model. Mechanistically, this effect occurs because perforin increases BSCB permeability, causing inflammatory cells and related cytokines to infiltrate and disrupt the nervous system.
Collapse
Affiliation(s)
- Zhaoxiang Liu
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China; Department of Neurosurgery, Xiangtan Central Hospital, 120 Heping RD, XiangTan Hunan, 411100, China
| | - Hua Zhang
- Biomedical Translation Research Institute, Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China
| | - Hong Xia
- Department of Orthopedics, Xiangtan Central Hospital, 120 Heping RD, XiangTan Hunan, 411100, China
| | - Baocheng Wang
- Shenzhen Graduate School of Tsinghua University, 2279 Lishui RD, Nanshan District, Shenzhen, 518055, China
| | - Renwen Zhang
- Chinese Traditional Medicine School, Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China
| | - Qun Zeng
- Department of Neurosurgery, Xiangtan Central Hospital, 120 Heping RD, XiangTan Hunan, 411100, China
| | - Lingzhi Guo
- Department of Neurosurgery, Xiangtan Central Hospital, 120 Heping RD, XiangTan Hunan, 411100, China
| | - Kui Shen
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China
| | - BaTa Wang
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China
| | - Yanheng Zhong
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China
| | - Zhizhong Li
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China.
| | - Guodong Sun
- Department of Orthopedics, First Affiliated Hospital of Jinan University, 613 West Huangpu RD, Guangzhou, 510632, China.
| |
Collapse
|
20
|
Shen HY, Huang N, Reemmer J, Xiao L. Adenosine Actions on Oligodendroglia and Myelination in Autism Spectrum Disorder. Front Cell Neurosci 2018; 12:482. [PMID: 30581380 PMCID: PMC6292987 DOI: 10.3389/fncel.2018.00482] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/26/2018] [Indexed: 11/21/2022] Open
Abstract
Autism spectrum disorder (ASD) is the most commonly diagnosed neurodevelopmental disorder. Independent of neuronal dysfunction, ASD and its associated comorbidities have been linked to hypomyelination and oligodendroglial dysfunction. Additionally, the neuromodulator adenosine has been shown to affect certain ASD comorbidities and symptoms, such as epilepsy, impairment of cognitive function, and anxiety. Adenosine is both directly and indirectly responsible for regulating the development of oligodendroglia and myelination through its interaction with, and modulation of, several neurotransmitters, including glutamate, dopamine, and serotonin. In this review, we will focus on the recent discoveries in adenosine interaction with physiological and pathophysiological activities of oligodendroglia and myelination, as well as ASD-related aspects of adenosine actions on neuroprotection and neuroinflammation. Moreover, we will discuss the potential therapeutic value and clinical approaches of adenosine manipulation against hypomyelination in ASD.
Collapse
Affiliation(s)
- Hai-Ying Shen
- Robert Stone Dow Neurobiology Department, Legacy Research Institute, Legacy Health, Portland, OR, United States.,Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, United States
| | - Nanxin Huang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jesica Reemmer
- Robert Stone Dow Neurobiology Department, Legacy Research Institute, Legacy Health, Portland, OR, United States
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
21
|
Akbari A, Khalili-Fomeshi M, Ashrafpour M, Moghadamnia AA, Ghasemi-Kasman M. Adenosine A 2A receptor blockade attenuates spatial memory deficit and extent of demyelination areas in lyolecithin-induced demyelination model. Life Sci 2018; 205:63-72. [PMID: 29730168 DOI: 10.1016/j.lfs.2018.05.007] [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: 02/11/2018] [Revised: 04/21/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022]
Abstract
In recent years, inactivation of A2A adenosine receptors has been emerged as a novel strategy for treatment of several neurodegenerative diseases. Although numerous studies have shown the beneficial effects of A2A receptors blockade on spatial memory, the impacts of selective adenosine A2A receptors on memory performance has not yet been examined in the context of demyelination. In the present study, we evaluated the effect of A2A receptor antagonist SCH58261 on spatial memory and myelination in an experimental model of focal demyelination in rat fimbria. Demyelination was induced by local injection of lysolecithin (LPC) 1% (2 μl) into the hippocampus fimbria. SCH58261 (20 μg/0.5 μl or 40 μg/0.5 μl) was daily injected intracerebroventricularly (i.c.v.) for 10 days post LPC injection. The Morris water maze test was used to assess the spatial learning and memory on day 6 post lesion. Myelin staining and immunostaining against astrocytes/microglia were carried out 10 days post LPC injection. The administration of adenosine A2A receptor antagonist prevented the spatial memory impairment in LPC receiving animals. Myelin staining revealed that application of SCH58261 reduces the extent of demyelination areas in the fimbria. Furthermore, the level of astrocytes and microglia activation was attenuated following administration of A2A receptor antagonist. Collectively, the results of this study suggest that A2A receptor blockade can improve the spatial memory and protect myelin sheath, which might be considered as a novel therapeutic approach for multiple sclerosis disease.
Collapse
Affiliation(s)
- Atefeh Akbari
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | - Manouchehr Ashrafpour
- Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Physiology, Faculty of Medical Sciences, Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Pharmacology, Faculty of Medical Sciences, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
22
|
Sobrido-Cameán D, Barreiro-Iglesias A. Role of Caspase-8 and Fas in Cell Death After Spinal Cord Injury. Front Mol Neurosci 2018; 11:101. [PMID: 29666570 PMCID: PMC5891576 DOI: 10.3389/fnmol.2018.00101] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/15/2018] [Indexed: 01/10/2023] Open
Abstract
Spinal cord injury (SCI) causes the death of neurons and glial cells due to the initial mechanical forces (i.e., primary injury) and through a cascade of secondary molecular events (e.g., inflammation or excitotoxicity) that exacerbate cell death. The loss of neurons and glial cells that are not replaced after the injury is one of the main causes of disability after SCI. Evidence accumulated in last decades has shown that the activation of apoptotic mechanisms is one of the factors causing the death of intrinsic spinal cord (SC) cells following SCI. Although this is not as clear for brain descending neurons, some studies have also shown that apoptosis can be activated in the brain following SCI. There are two main apoptotic pathways, the extrinsic and the intrinsic pathways. Activation of caspase-8 is an important step in the initiation of the extrinsic pathway. Studies in rodents have shown that caspase-8 is activated in SC glial cells and neurons and that the Fas receptor plays a key role in its activation following a traumatic SCI. Recent work in the lamprey model of SCI has also shown the retrograde activation of caspase-8 in brain descending neurons following SCI. Here, we review our current knowledge on the role of caspase-8 and the Fas pathway in cell death following SCI. We also provide a perspective for future work on this process, like the importance of studying the possible contribution of Fas/caspase-8 signaling in the degeneration of brain neurons after SCI in mammals.
Collapse
Affiliation(s)
- Daniel Sobrido-Cameán
- Department of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Antón Barreiro-Iglesias
- Department of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
23
|
Acton D, Miles GB. Gliotransmission and adenosinergic modulation: insights from mammalian spinal motor networks. J Neurophysiol 2017; 118:3311-3327. [PMID: 28954893 DOI: 10.1152/jn.00230.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Astrocytes are proposed to converse with neurons at tripartite synapses, detecting neurotransmitter release and responding with release of gliotransmitters, which in turn modulate synaptic strength and neuronal excitability. However, a paucity of evidence from behavioral studies calls into question the importance of gliotransmission for the operation of the nervous system in healthy animals. Central pattern generator (CPG) networks in the spinal cord and brain stem coordinate the activation of muscles during stereotyped activities such as locomotion, inspiration, and mastication and may therefore provide tractable models in which to assess the contribution of gliotransmission to behaviorally relevant neural activity. We review evidence for gliotransmission within spinal locomotor networks, including studies indicating that adenosine derived from astrocytes regulates the speed of locomotor activity via metamodulation of dopamine signaling.
Collapse
Affiliation(s)
- David Acton
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife , United Kingdom
| | - Gareth B Miles
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife , United Kingdom
| |
Collapse
|
24
|
Liu X, Ma L, Zhang S, Ren Y, Dirksen RT. CD73 Controls Extracellular Adenosine Generation in the Trigeminal Nociceptive Nerves. J Dent Res 2017; 96:671-677. [PMID: 28530470 DOI: 10.1177/0022034517692953] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purinergic signaling is involved in pain generation and modulation in the nociceptive sensory nervous system. Adenosine triphosphate (ATP) induces pain via activation of ionotropic P2X receptors while adenosine mediates analgesia via activation of metabotropic P1 receptors. These purinergic signaling are determined by ecto-nucleotidases that control ATP degradation and adenosine generation. Using enzymatic histochemistry, we detected ecto-AMPase activity in dental pulp, trigeminal ganglia (TG) neurons, and their nerve fibers. Using immunofluorescence staining, we confirmed the expression of ecto-5'-nucleotidase (CD73) in trigeminal nociceptive neurons and their axonal fibers, including the nociceptive nerve fibers projecting into the brainstem. In addition, we detected the existence of CD73 and ecto-AMPase activity in the nociceptive lamina of the trigeminal subnucleus caudalis (TSNC) in the brainstem. Furthermore, we demonstrated that incubation with specific anti-CD73 serum significantly reduced the ecto-AMPase activity in the nociceptive lamina in the brainstem. Our results indicate that CD73 might participate in nociceptive modulation by affecting extracellular adenosine generation in the trigeminal nociceptive pathway. Disruption of TG neuronal ecto-nucleotidase expression and axonal terminal localization under certain circumstances such as chronic inflammation, oxidant stress, local constriction, and injury in trigeminal nerves may contribute to the pathogenesis of orofacial neuropathic pain.
Collapse
Affiliation(s)
- X Liu
- 1 Department of Dentistry, Eastman Institute for Oral Health, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,2 Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - L Ma
- 1 Department of Dentistry, Eastman Institute for Oral Health, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,3 Department of Dentistry, School of Stomatology, Zhengzhou University, Zhengzhou, China
| | - S Zhang
- 1 Department of Dentistry, Eastman Institute for Oral Health, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Y Ren
- 1 Department of Dentistry, Eastman Institute for Oral Health, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - R T Dirksen
- 2 Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
25
|
Wei X, Zhou Z, Li L, Gu J, Wang C, Xu F, Dong Q, Zhou X. Intrathecal Injection of 3-Methyladenine Reduces Neuronal Damage and Promotes Functional Recovery via Autophagy Attenuation after Spinal Cord Ischemia/Reperfusion Injury in Rats. Biol Pharm Bull 2017; 39:665-73. [PMID: 27150140 DOI: 10.1248/bpb.b15-00610] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study aimed to determine the occurrence of autophagy following ischemia/reperfusion (I/R) injury in the rat spinal cord and whether autophagy inhibition contributes to neural tissue damage and locomotor impairment. A spinal cord I/R model was induced via descending thoracic aorta occlusion for 10 min using systemic hypotension (40 mmHg) in adult male Sprague-Dawley rats. Then, 600 nmol 3-methyladenine (3-MA) or vehicle was intrathecally administered. Ultrastructural spinal cord changes were observed via transmission electron microscopy (TEM) and immunofluorescent double-labeling. Western blots were used to determine the protein expression of microtubule-associated protein light chain 3 (LC3) and Beclin 1. Autophagy was activated after spinal cord I/R injury as demonstrated by significantly increased LC3 and Beclin 1 expression at 3-48 h after injury. Furthermore, TEM images indicated the presence of autophagosomes and autolysosomes in the injured spinal cord. 3-MA significantly decreased LC3 and Beclin 1 expression and the number of LC3-positive cells in spinal cord of I/R versus vehicle groups. Moreover, the 3-MA-treated rats exhibited better neurobehavioral scores compared with control rats. These findings suggest activation of autophagy leading to neuronal cell death in the I/R injured spinal cord. These effects were significantly inhibited by intrathecal 3-MA administration. Thus intrathecal 3-MA administration may represent a novel treatment target following spinal cord I/R injury.
Collapse
Affiliation(s)
- Xing Wei
- Department of Anesthesia, the Second Affiliated Hospital of Soochow University
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Adenosine receptor targets for pain. Neuroscience 2016; 338:1-18. [DOI: 10.1016/j.neuroscience.2015.10.031] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/29/2015] [Accepted: 10/15/2015] [Indexed: 12/21/2022]
|
27
|
Immune Surveillance of the CNS following Infection and Injury. Trends Immunol 2016; 36:637-650. [PMID: 26431941 DOI: 10.1016/j.it.2015.08.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/10/2015] [Accepted: 08/10/2015] [Indexed: 12/24/2022]
Abstract
The central nervous system (CNS) contains a sophisticated neural network that must be constantly surveyed in order to detect and mitigate a diverse array of challenges. The innate and adaptive immune systems actively participate in this surveillance, which is critical for the maintenance of CNS homeostasis and can facilitate the resolution of infections, degeneration, and tissue damage. Infections and sterile injuries represent two common challenges imposed on the CNS that require a prompt immune response. While the inducers of these two challenges differ in origin, the resultant responses orchestrated by the CNS share some overlapping features. Here, we review how the CNS immunologically discriminates between pathogens and sterile injuries, mobilizes an immune reaction, and, ultimately, regulates local and peripherally-derived immune cells to provide a supportive milieu for tissue repair.
Collapse
|
28
|
Ingwersen J, Wingerath B, Graf J, Lepka K, Hofrichter M, Schröter F, Wedekind F, Bauer A, Schrader J, Hartung HP, Prozorovski T, Aktas O. Dual roles of the adenosine A2a receptor in autoimmune neuroinflammation. J Neuroinflammation 2016; 13:48. [PMID: 26920550 PMCID: PMC4768407 DOI: 10.1186/s12974-016-0512-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/16/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Conditions of inflammatory tissue distress are associated with high extracellular levels of adenosine, due to increased adenosine triphosphate (ATP) degradation upon cellular stress or the release of extracellular ATP upon cell death, which can be degraded to adenosine by membrane-bound ecto-enzymes like CD39 and CD73. Adenosine is recognised to mediate anti-inflammatory effects via the adenosine A2a receptor (A2aR), as shown in experimental models of arthritis. Here, using pharmacological interventions and genetic inactivation, we investigated the roles of A2aR in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). METHODS We used two independent mouse EAE variants, i.e. active immunization in C57BL/6 with myelin oligodendrocyte glycoprotein (MOG)35-55 or transfer-EAE by proteolipid protein (PLP)139-155-stimulated T lymphocytes and EAE in mice treated with A2aR-agonist CGS21680 at different stages of disease course and in mice lacking A2aR (A2aR(-/-)) compared to direct wild-type littermates. In EAE, we analysed myelin-specific proliferation and cytokine synthesis ex vivo, as well as inflammation and demyelination by immunohistochemistry. In vitro, we investigated the effect of A2aR on migration of CD4(+) T cells, macrophages and microglia, as well as the impact of A2aR on phagocytosis of macrophages and microglia. Statistical tests were Mann-Whitney U and Student's t test. RESULTS We found an upregulation of A2aR in the central nervous system (CNS) in EAE, predominantly detected on T cells and macrophages/microglia within the inflamed tissue. Preventive EAE treatment with A2aR-specific agonist inhibited myelin-specific T cell proliferation ex vivo and ameliorated disease, while application of the same agonist after disease onset exacerbated non-remitting EAE progression and resulted in more severe tissue destruction. Accordingly, A2aR-deficient mice showed accelerated and exacerbated disease manifestation with increased frequencies of IFN-γ-, IL-17- and GM-CSF-producing CD4(+) T helper cells and higher numbers of inflammatory lesions in the early stage. However, EAE quickly ameliorated and myelin debris accumulation was lower in A2aR(-/-) mice. In vitro, activation of A2aR inhibited phagocytosis of myelin by macrophages and primary microglia as well as migration of CD4(+) T cells, macrophages and primary microglia. CONCLUSIONS A2aR activation exerts a complex pattern in chronic autoimmune neurodegeneration: while providing anti-inflammatory effects on T cells and thus protection at early stages, A2aR seems to play a detrimental role during later stages of disease and may thus contribute to sustained tissue damage within the inflamed CNS.
Collapse
Affiliation(s)
- J Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - B Wingerath
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - J Graf
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - K Lepka
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - M Hofrichter
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - F Schröter
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany. .,Current address: Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, HeinrichHeine University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - F Wedekind
- Institute of Neuroscience and Medicine, INM-2, Research Center Jülich, Leo-Brandt-Str., 52425, Jülich, Germany.
| | - A Bauer
- Institute of Neuroscience and Medicine, INM-2, Research Center Jülich, Leo-Brandt-Str., 52425, Jülich, Germany.
| | - J Schrader
- Cardiovascular Physiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany.
| | - H-P Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - T Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - O Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
29
|
van Tilborg E, Heijnen CJ, Benders MJ, van Bel F, Fleiss B, Gressens P, Nijboer CH. Impaired oligodendrocyte maturation in preterm infants: Potential therapeutic targets. Prog Neurobiol 2015; 136:28-49. [PMID: 26655283 DOI: 10.1016/j.pneurobio.2015.11.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 11/02/2015] [Accepted: 11/18/2015] [Indexed: 12/20/2022]
Abstract
Preterm birth is an evolving challenge in neonatal health care. Despite declining mortality rates among extremely premature neonates, morbidity rates remain very high. Currently, perinatal diffuse white matter injury (WMI) is the most commonly observed type of brain injury in preterm infants and has become an important research area. Diffuse WMI is associated with impaired cognitive, sensory and psychological functioning and is increasingly being recognized as a risk factor for autism-spectrum disorders, ADHD, and other psychological disturbances. No treatment options are currently available for diffuse WMI and the underlying pathophysiological mechanisms are far from being completely understood. Preterm birth is associated with maternal inflammation, perinatal infections and disrupted oxygen supply which can affect the cerebral microenvironment by causing activation of microglia, astrogliosis, excitotoxicity, and oxidative stress. This intricate interplay of events negatively influences oligodendrocyte development, causing arrested oligodendrocyte maturation or oligodendrocyte cell death, which ultimately results in myelination failure in the developing white matter. This review discusses the current state in perinatal WMI research, ranging from a clinical perspective to basic molecular pathophysiology. The complex regulation of oligodendrocyte development in healthy and pathological conditions is described, with a specific focus on signaling cascades that may play a role in WMI. Furthermore, emerging concepts in the field of WMI and issues regarding currently available animal models are put forward. Novel insights into the molecular mechanisms underlying impeded oligodendrocyte maturation in diffuse WMI may aid the development of novel treatment options which are desperately needed to improve the quality-of-life of preterm neonates.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manon J Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van Bel
- Department of Neonatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bobbi Fleiss
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Pierre Gressens
- Inserm, Paris U1141, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS, Paris 1141, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
30
|
Mohamed RA, Agha AM, Abdel-Rahman AA, Nassar NN. Role of adenosine A2A receptor in cerebral ischemia reperfusion injury: Signaling to phosphorylated extracellular signal-regulated protein kinase (pERK1/2). Neuroscience 2015; 314:145-59. [PMID: 26642806 DOI: 10.1016/j.neuroscience.2015.11.059] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/02/2015] [Accepted: 11/25/2015] [Indexed: 12/11/2022]
Abstract
Following brain ischemia reperfusion (IR), the dramatic increase in adenosine activates A2AR to induce further neuronal damage. Noteworthy, A2A antagonists have proven efficacious in halting IR injury, however, the detailed downstream signaling remains elusive. To this end, the present study aimed to investigate the possible involvement of phospho-extracellular signal-regulated kinase (pERK1/2) pathway in mediating protection afforded by the central A2A blockade. Male Wistar rats (250-270 g) subjected to bilateral carotid occlusion for 45 min followed by a 24-h reperfusion period showed increased infarct size corroborating histopathological damage, memory impairment and motor incoordination as well as increased locomotor activity. Those events were mitigated by the unilateral intrahippocampal administration of the selective A2A antagonist SCH58261 via a decrease in pERK1/2 downstream from diacyl glycerol (DAG) signaling. Consequent to pERK1/2 inhibition, reduced hippocampal microglial activation, glial tumor necrosis factor-alpha (TNF-α) and brain-derived neurotropic factor (BDNF) expression, glutamate (Glu), inducible nitric oxide synthase (iNOS) and thiobarbituric acid reactive substances (TBARS) were evident in animals receiving SCH58261. Additionally, the anti-inflammatory cytokine interleukin-10 (IL-10) increased following nuclear factor (erythroid-derived 2)-like 2 (Nrf-2). Taken all together, these events suppressed apoptotic pathways via a reduction in cytochrome c (Cyt. c) as well as caspase-3 supporting a crucial role for pERK1/2 inhibition in consequent reduction of inflammatory and excitotoxic cascades as well as correction of the redox imbalance.
Collapse
Affiliation(s)
- R A Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - A M Agha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - A A Abdel-Rahman
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, NC, USA.
| | - N N Nassar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
31
|
Pedata F, Dettori I, Coppi E, Melani A, Fusco I, Corradetti R, Pugliese AM. Purinergic signalling in brain ischemia. Neuropharmacology 2015; 104:105-30. [PMID: 26581499 DOI: 10.1016/j.neuropharm.2015.11.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 12/18/2022]
Abstract
Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia a primary damage due to the early massive increase of extracellular glutamate is followed by activation of resident immune cells, i.e microglia, and production or activation of inflammation mediators. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. Extracellular concentrations of ATP and adenosine in the brain increase dramatically during ischemia in concentrations able to stimulate their respective specific P2 and P1 receptors. Both ATP P2 and adenosine P1 receptor subtypes exert important roles in ischemia. Although adenosine exerts a clear neuroprotective effect through A1 receptors during ischemia, the use of selective A1 agonists is hampered by undesirable peripheral effects. Evidence up to now in literature indicate that A2A receptor antagonists provide protection centrally by reducing excitotoxicity, while agonists at A2A (and possibly also A2B) and A3 receptors provide protection by controlling massive infiltration and neuroinflammation in the hours and days after brain ischemia. Among P2X receptors most evidence indicate that P2X7 receptor contribute to the damage induced by the ischemic insult due to intracellular Ca(2+) loading in central cells and facilitation of glutamate release. Antagonism of P2X7 receptors might represent a new treatment to attenuate brain damage and to promote proliferation and maturation of brain immature resident cells that can promote tissue repair following cerebral ischemia. Among P2Y receptors, antagonists of P2Y12 receptors are of value because of their antiplatelet activity and possibly because of additional anti-inflammatory effects. Moreover strategies that modify adenosine or ATP concentrations at injury sites might be of value to limit damage after ischemia. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy.
| | - Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Elisabetta Coppi
- Department of Health Sciences, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Alessia Melani
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Irene Fusco
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| |
Collapse
|
32
|
Acton D, Miles GB. Stimulation of Glia Reveals Modulation of Mammalian Spinal Motor Networks by Adenosine. PLoS One 2015; 10:e0134488. [PMID: 26252389 PMCID: PMC4529192 DOI: 10.1371/journal.pone.0134488] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 07/09/2015] [Indexed: 01/05/2023] Open
Abstract
Despite considerable evidence that glia can release modulators to influence the excitability of neighbouring neurons, the importance of gliotransmission for the operation of neural networks and in shaping behaviour remains controversial. Here we characterise the contribution of glia to the modulation of the mammalian spinal central pattern generator for locomotion, the output of which is directly relatable to a defined behaviour. Glia were stimulated by specific activation of protease-activated receptor-1 (PAR1), an endogenous G-protein coupled receptor preferentially expressed by spinal glia during ongoing activity of the spinal central pattern generator for locomotion. Selective activation of PAR1 by the agonist TFLLR resulted in a reversible reduction in the frequency of locomotor-related bursting recorded from ventral roots of spinal cord preparations isolated from neonatal mice. In the presence of the gliotoxins methionine sulfoximine or fluoroacetate, TFLLR had no effect, confirming the specificity of PAR1 activation to glia. The modulation of burst frequency upon PAR1 activation was blocked by the non-selective adenosine-receptor antagonist theophylline and by the A1-receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine, but not by the A2A-receptor antagonist SCH5826, indicating production of extracellular adenosine upon glial stimulation, followed by A1-receptor mediated inhibition of neuronal activity. Modulation of network output following glial stimulation was also blocked by the ectonucleotidase inhibitor ARL67156, indicating glial release of ATP and its subsequent degradation to adenosine rather than direct release of adenosine. Glial stimulation had no effect on rhythmic activity recorded following blockade of inhibitory transmission, suggesting that glial cell-derived adenosine acts via inhibitory circuit components to modulate locomotor-related output. Finally, the modulation of network output by endogenous adenosine was found to scale with the frequency of network activity, implying activity-dependent release of adenosine. Together, these data indicate that glia play an active role in the modulation of mammalian locomotor networks, providing negative feedback control that may stabilise network activity.
Collapse
Affiliation(s)
- David Acton
- School of Psychology and Neuroscience, University of St Andrews, Fife, United Kingdom
| | - Gareth B. Miles
- School of Psychology and Neuroscience, University of St Andrews, Fife, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Navarrete-Opazo A, Vinit S, Dougherty BJ, Mitchell GS. Daily acute intermittent hypoxia elicits functional recovery of diaphragm and inspiratory intercostal muscle activity after acute cervical spinal injury. Exp Neurol 2015; 266:1-10. [PMID: 25687551 PMCID: PMC4716671 DOI: 10.1016/j.expneurol.2015.02.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/14/2015] [Accepted: 02/06/2015] [Indexed: 01/31/2023]
Abstract
A major cause of mortality after spinal cord injury is respiratory failure. In normal rats, acute intermittent hypoxia (AIH) induces respiratory motor plasticity, expressed as diaphragm (Dia) and second external intercostal (T2 EIC) long-term facilitation (LTF). Dia (not T2 EIC) LTF is enhanced by systemic adenosine 2A (A2A) receptor inhibition in normal rats. We investigated the respective contributions of Dia and T2 EIC to daily AIH-induced functional recovery of breathing capacity with/without A2A receptor antagonist (KW6002, i.p.) following C2 hemisection (C2HS). Rats received daily AIH (dAIH: 10, 5-min episodes, 10.5% O2; 5-min normoxic intervals; 7 successive days beginning 7days post-C2HS) or daily normoxia (dNx) with/without KW6002, followed by weekly (reminder) presentations for 8weeks. Ventilation and EMGs from bilateral diaphragm and T2 EIC muscles were measured with room air breathing (21% O2) and maximum chemoreceptor stimulation ( MCS 7% CO2, 10.5% O2). dAIH increased tidal volume (VT) in C2HS rats breathing room air (dAIH+vehicle: 0.47±0.02, dNx+vehicle: 0.40±0.01ml/100g; p<0.05) and MCS (dAIH+vehicle: 0.83±0.01, dNx+vehicle: 0.73±0.01ml/100g; p<0.001); KW6002 had no significant effect. dAIH enhanced contralateral (uninjured) diaphragm EMG activity, an effect attenuated by KW6002, during room air breathing and MCS (p<0.05). Although dAIH enhanced contralateral T2 EIC EMG activity during room air breathing, KW6002 had no effect. dAIH had no statistically significant effects on diaphragm or T2 EIC EMG activity ipsilateral to injury. Thus, two weeks post-C2HS: 1) dAIH enhances breathing capacity by effects on contralateral diaphragm and T2 EIC activity; and 2) dAIH-induced recovery is A2A dependent in diaphragm, but not T2 EIC. Daily AIH may be a useful in promoting functional recovery of breathing capacity after cervical spinal injury, but A2A receptor antagonists (e.g. caffeine) may undermine its effectiveness shortly after injury.
Collapse
Affiliation(s)
- A Navarrete-Opazo
- Department of Comparative Biosciences University of Wisconsin-Madison, Madison, WI 53706, USA
| | - S Vinit
- Department of Comparative Biosciences University of Wisconsin-Madison, Madison, WI 53706, USA
| | - B J Dougherty
- Department of Comparative Biosciences University of Wisconsin-Madison, Madison, WI 53706, USA
| | - G S Mitchell
- Department of Comparative Biosciences University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
34
|
Liu X, Li C, Liang F, Wang Y, Li Z, Yang J. Effects of hyperbaric oxygen on glucose-regulated protein 78 and c-Jun N-terminal kinase expression after spinal cord injury in rats. Int J Clin Exp Med 2015; 8:3309-3317. [PMID: 26064220 PMCID: PMC4443054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/25/2015] [Indexed: 06/04/2023]
Abstract
Spinal cord injury (SCI) is not only devastating but also represents a public health burden for society. Endoplasmic reticulum stress (ERS) is implicated in secondary injury following damage to the SC. Hyperbaric oxygen (HBO) treatment can improve the recovery of motor function after SCI, but the effect of HBO on the ERS response is unknown. This study tested the hypothesis that HBO treatment protects against secondary SCI by inhibiting the ERS response via regulation of glucose-regulated protein (GRP) 78 and c-Jun N-terminal kinase (JNK) expression. Rats were randomly assigned to sham, SCI, and SCI + HBO groups and the extent of neuronal damage and neurological recovery were evaluated 1, 3, 7, and 14 days after surgery. GRP78 and JNK expression was evaluated by immunohistochemical, western blot, and real-time reverse transcription-polymerase chain reaction analyses, while caspase-3 activation was evaluated by enzyme-linked immunosorbent assay. SCI resulted in an upregulation in GRP78 and JNK expression compared to sham-operated animals. HBO treatment increased GRP78 level, but decreased that of JNK and suppressed caspase-3 activation as well as neuronal damage relative to the SCI group. In addition, hind limb motor function was improved by HBO treatment. HBO treatment reduces SCI-induced neuronal death and promotes the recovery of neurological function recovery by inhibiting the ERS response via modulation of GRP78 and JNK expression levels.
Collapse
Affiliation(s)
- Xuehua Liu
- Department of Hyperbaric Oxygen, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing 100020, China
| | - Chunsheng Li
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing 100020, China
| | - Fang Liang
- Department of Hyperbaric Oxygen, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing 100020, China
| | - Yong Wang
- Department of Hyperbaric Oxygen, Beijing Fuxing Hospital, Capital Medical UniversityBeijing 100038, China
| | - Zhuo Li
- Department of Hyperbaric Oxygen, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing 100020, China
| | - Jing Yang
- Department of Hyperbaric Oxygen, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing 100020, China
| |
Collapse
|
35
|
Adenosine A2A receptors modulate acute injury and neuroinflammation in brain ischemia. Mediators Inflamm 2014; 2014:805198. [PMID: 25165414 PMCID: PMC4138795 DOI: 10.1155/2014/805198] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/26/2014] [Accepted: 07/10/2014] [Indexed: 01/07/2023] Open
Abstract
The extracellular concentration of adenosine in the brain increases dramatically during ischemia. Adenosine A2A receptor is expressed in neurons and glial cells and in inflammatory cells (lymphocytes and granulocytes). Recently, adenosine A2A receptor emerged as a potential therapeutic attractive target in ischemia. Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia the early massive increase of extracellular glutamate is followed by activation of resident immune cells, that is, microglia, and production or activation of inflammation mediators. Proinflammatory cytokines, which upregulate cell adhesion molecules, exert an important role in promoting recruitment of leukocytes that in turn promote expansion of the inflammatory response in ischemic tissue. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. A2A receptors present on central cells and on blood cells account for important effects depending on the time-related evolution of the pathological condition. Evidence suggests that A2A receptor antagonists provide early protection via centrally mediated control of excessive excitotoxicity, while A2A receptor agonists provide protracted protection by controlling massive blood cell infiltration in the hours and days after ischemia. Focus on inflammatory responses provides for adenosine A2A receptor agonists a wide therapeutic time-window of hours and even days after stroke.
Collapse
|
36
|
Zheng X, Zhang X, Kang A, Ran C, Wang G, Hao H. Thinking outside the brain for cognitive improvement: Is peripheral immunomodulation on the way? Neuropharmacology 2014; 96:94-104. [PMID: 24978103 DOI: 10.1016/j.neuropharm.2014.06.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 06/16/2014] [Accepted: 06/19/2014] [Indexed: 02/07/2023]
Abstract
Cognitive impairment is a devastating condition commonly observed with normal aging and neurodegenerative disorders such as Alzheimer's Disease (AD). Although major efforts to prevent or slow down cognitive decline are largely focused within the central nervous system (CNS), it has become clear that signals from the systemic milieu are closely associated with the dysfunctional brain. In particular, the bidirectional crosstalk between the CNS and peripheral immune system plays a decisive role in shaping neuronal survival and function via neuroimmune, neuroendocrinal and bioenergetic mechanisms. Importantly, it is emerging that some neuroprotective and cognition-strengthening drugs may work by targeting the brain-periphery interactions, which could be intriguingly achieved without entering the CNS. We describe here how recent advances in dissecting cognitive deficits from a systems-perspective have contributed to a non-neurocentric understanding of its pathogenesis and treatment strategy. We also discuss the therapeutic and diagnostic implications of these exciting progresses and consider some key issues in the clinical translation. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Xiao Zheng
- Nanjing University of Chinese Medicine Affiliated Hospital, Nanjing 210029, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Xueli Zhang
- Zhong Da Hospital, Southeast University, Nanjing 210009, China
| | - An Kang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chongzhao Ran
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston 02129, United States
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
37
|
Smith MD, Bhatt DP, Geiger JD, Rosenberger TA. Acetate supplementation modulates brain adenosine metabolizing enzymes and adenosine A₂A receptor levels in rats subjected to neuroinflammation. J Neuroinflammation 2014; 11:99. [PMID: 24898794 PMCID: PMC4050445 DOI: 10.1186/1742-2094-11-99] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 05/19/2014] [Indexed: 12/20/2022] Open
Abstract
Background Acetate supplementation reduces neuroglia activation and pro-inflammatory cytokine expression in rat models of neuroinflammation and Lyme neuroborreliosis. Because single-dose glyceryl triacetate (GTA) treatment increases brain phosphocreatine and reduces brain AMP levels, we postulate that GTA modulates adenosine metabolizing enzymes and receptors, which may be a possible mechanism to reduce neuroinflammation. Methods To test this hypothesis, we quantified the ability of GTA to alter brain levels of ecto-5’-nucleotidase (CD73), adenosine kinase (AK), and adenosine A2A receptor using western blot analysis and CD73 activity by measuring the rate of AMP hydrolysis. Neuroinflammation was induced by continuous bacterial lipopolysaccharide (LPS) infusion in the fourth ventricle of the brain for 14 and 28 days. Three treatment strategies were employed, one and two where rats received prophylactic GTA through oral gavage with LPS infusion for 14 or 28 days. In the third treatment regimen, an interventional strategy was used where rats were subjected to 28 days of neuroinflammation, and GTA treatment was started on day 14 following the start of the LPS infusion. Results We found that rats subjected to neuroinflammation for 28 days had a 28% reduction in CD73 levels and a 43% increase in AK levels that was reversed with prophylactic acetate supplementation. CD73 activity in these rats was increased by 46% with the 28-day GTA treatment compared to the water-treated rats. Rats subjected to neuroinflammation for 14 days showed a 50% increase in levels of the adenosine A2A receptor, which was prevented with prophylactic acetate supplementation. Interventional GTA therapy, beginning on day 14 following the induction of neuroinflammation, resulted in a 67% increase in CD73 levels and a 155% increase in adenosine A2A receptor levels. Conclusion These results support the hypothesis that acetate supplementation can modulate brain CD73, AK and adenosine A2A receptor levels, and possibly influence purinergic signaling.
Collapse
Affiliation(s)
| | | | | | - Thad A Rosenberger
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Grand Forks, North Dakota 58203, USA.
| |
Collapse
|
38
|
Using caffeine and other adenosine receptor antagonists and agonists as therapeutic tools against neurodegenerative diseases: a review. Life Sci 2014; 101:1-9. [PMID: 24530739 DOI: 10.1016/j.lfs.2014.01.083] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 01/22/2014] [Accepted: 01/29/2014] [Indexed: 12/22/2022]
Abstract
Caffeine is the most consumed pychostimulant in the world, and it is known to affect basic and fundamental human processes such as sleep, arousal, cognition and learning and memory. It works as a nonselective blocker of adenosine receptors (A1, A2a, A2b and A3) and has been related to the regulation of heart rate, the contraction/relaxation of cardiac and smooth muscles, and the neural signaling in the central nervous system (CNS). Since the late 1990s, studies using adenosine receptor antagonists, such as Caffeine, to block the A1 and A2a adenosine receptor subtypes have shown to reduce the physical, cellular and molecular damages caused by a spinal cord injury (SCI) or a stroke (cerebral infarction) and by other neurodegenerative diseases such as Parkinson's and Alzheimer's diseases. Interestingly, other studies using adenosine receptor agonists have also shown to provide a neuroprotective effect on various models of neurodegenerative diseases through the reduction of excitatory neurotransmitter release, apoptosis and inflammatory responses, among others. The seemingly paradoxical use of both adenosine receptor agonists and antagonists as neuroprotective agents has been attributed to differences in dosage levels, drug delivery method, extracellular concentration of excitatory neurotransmitters and stage of disease progression. We discuss and compare recent findings using both antagonists and agonists of adenosine receptors in animal models and patients that have suffered spinal cord injuries, brain strokes, and Parkinson's and Alzheimer's diseases. Additionally, we propose alternative interpretations on the seemingly paradoxical use of these drugs as potential pharmacological tools to treat these various types of neurodegenerative diseases.
Collapse
|
39
|
|
40
|
Gaudin A, Yemisci M, Eroglu H, Lepêtre-Mouelhi S, Turkoglu OF, Dönmez-Demir B, Caban S, Fevzi Sargon M, Garcia-Argote S, Pieters G, Loreau O, Rousseau B, Tagit O, Hildebrandt N, Le Dantec Y, Mougin J, Valetti S, Chacun H, Nicolas V, Desmaële D, Andrieux K, Capan Y, Dalkara T, Couvreur P. Squalenoyl adenosine nanoparticles provide neuroprotection after stroke and spinal cord injury. NATURE NANOTECHNOLOGY 2014; 9:1054-1062. [PMID: 25420034 PMCID: PMC4351925 DOI: 10.1038/nnano.2014.274] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 10/21/2014] [Indexed: 05/19/2023]
Abstract
There is an urgent need to develop new therapeutic approaches for the treatment of severe neurological trauma, such as stroke and spinal cord injuries. However, many drugs with potential neuropharmacological activity, such as adenosine, are inefficient upon systemic administration because of their fast metabolization and rapid clearance from the bloodstream. Here, we show that conjugation of adenosine to the lipid squalene and the subsequent formation of nanoassemblies allows prolonged circulation of this nucleoside, providing neuroprotection in mouse stroke and rat spinal cord injury models. The animals receiving systemic administration of squalenoyl adenosine nanoassemblies showed a significant improvement of their neurologic deficit score in the case of cerebral ischaemia, and an early motor recovery of the hindlimbs in the case of spinal cord injury. Moreover, in vitro and in vivo studies demonstrated that the nanoassemblies were able to extend adenosine circulation and its interaction with the neurovascular unit. This Article shows, for the first time, that a hydrophilic and rapidly metabolized molecule such as adenosine may become pharmacologically efficient owing to a single conjugation with the lipid squalene.
Collapse
Affiliation(s)
- Alice Gaudin
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Müge Yemisci
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara 06100, Turkey
| | - Hakan Eroglu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| | - Sinda Lepêtre-Mouelhi
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Omer Faruk Turkoglu
- Department of Neurosurgery, Ankara Ataturk Research & Education Hospital, 06800 Bilkent Ankara, Turkey
| | - Buket Dönmez-Demir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara 06100, Turkey
| | - Seçil Caban
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| | - Mustafa Fevzi Sargon
- Department of Anatomy, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | | | - Grégory Pieters
- CEA Saclay, iBiTecS-S/SCBM, Labex LERMIT, 91191 Gif-sur-Yvette, France
| | - Olivier Loreau
- CEA Saclay, iBiTecS-S/SCBM, Labex LERMIT, 91191 Gif-sur-Yvette, France
| | - Bernard Rousseau
- CEA Saclay, iBiTecS-S/SCBM, Labex LERMIT, 91191 Gif-sur-Yvette, France
| | - Oya Tagit
- NanoBioPhotonics, Institut d’Electronique Fondamentale, University of Paris-Sud XI, 91405, Orsay Cedex, France
| | - Niko Hildebrandt
- NanoBioPhotonics, Institut d’Electronique Fondamentale, University of Paris-Sud XI, 91405, Orsay Cedex, France
| | - Yannick Le Dantec
- EA3544, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Julie Mougin
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Sabrina Valetti
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Hélène Chacun
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Valérie Nicolas
- Institut d’Innovation Thérapeutique, IFR141 ITFM, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Didier Desmaële
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
| | - Karine Andrieux
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
- Correspondence and requests for materials should be adressed to P.C. and K.A. ,
| | - Yilmaz Capan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara 06100, Turkey
| | - Patrick Couvreur
- Institut Galien Paris-Sud UMR CNRS 8612, Faculty of Pharmacy, University of Paris-Sud XI, 92296 Châtenay-Malabry, France
- Correspondence and requests for materials should be adressed to P.C. and K.A. ,
| |
Collapse
|
41
|
de Lera Ruiz M, Lim YH, Zheng J. Adenosine A2A Receptor as a Drug Discovery Target. J Med Chem 2013; 57:3623-50. [DOI: 10.1021/jm4011669] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Manuel de Lera Ruiz
- Department
of Chemical Research, Merck Research Laboratories, 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Yeon-Hee Lim
- Department
of Chemical Research, Merck Research Laboratories, 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Junying Zheng
- Department
of Chemical Research, Merck Research Laboratories, 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| |
Collapse
|
42
|
Intrathecal injection of adenosine 2A receptor agonists reversed neuropathic allodynia through protein kinase (PK)A/PKC signaling. Brain Behav Immun 2013; 33:112-22. [PMID: 23811314 PMCID: PMC4447865 DOI: 10.1016/j.bbi.2013.06.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 06/03/2013] [Accepted: 06/21/2013] [Indexed: 01/05/2023] Open
Abstract
A single intrathecal dose of adenosine 2A receptor (A2AR) agonist was previously reported to produce a multi-week reversal of allodynia in a chronic constriction injury (CCI) model of neuropathic pain. We aimed to determine if this long-term reversal was induced by A2AR agonism versus more generalized across adenosine receptor subtypes, and begin to explore the intracellular signaling cascades involved. In addition, we sought to identify whether the enduring effect could be extended to other models of neuropathic pain. We tested an A1R and A2BR agonist in CCI and found the same long duration effect with A2BR but not A1R agonism. An A2AR agonist (ATL313) produced a significant long-duration reversal of mechanical allodynia induced by long established CCI (administered 6 weeks after surgery), spinal nerve ligation and sciatic inflammatory neuropathy. To determine if ATL313 had a direct effect on glia, ATL313 was coadministered with lipopolysaccharide to neonatal microglia and astrocytes in vitro. ATL313 significantly attenuated TNFα production in both microglia and astrocytes but had no effect on LPS induced IL-10. Protein kinase C significantly reversed the ATL313 effects on TNFα in vitro in microglia and astrocytes, while a protein kinase A inhibitor only effected microglia. Both intrathecal PKA and PKC inhibitors significantly reversed the effect of the A2AR agonist on neuropathic allodynia. Therefore, A2AR agonists administered IT remain an exciting novel target for the treatment of neuropathic pain.
Collapse
|
43
|
Kertmen H, Gürer B, Yılmaz ER, Şanlı AM, Sorar M, Arıkök AT, Sargon MF, Kanat MA, Ergüder BI, Şekerci Z. The protective effect of low-dose methotrexate on ischemia–reperfusion injury of the rabbit spinal cord. Eur J Pharmacol 2013; 714:148-56. [DOI: 10.1016/j.ejphar.2013.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/01/2013] [Accepted: 05/11/2013] [Indexed: 11/27/2022]
|
44
|
Gomes C, Ferreira R, George J, Sanches R, Rodrigues DI, Gonçalves N, Cunha RA. Activation of microglial cells triggers a release of brain-derived neurotrophic factor (BDNF) inducing their proliferation in an adenosine A2A receptor-dependent manner: A2A receptor blockade prevents BDNF release and proliferation of microglia. J Neuroinflammation 2013; 10:16. [PMID: 23363775 PMCID: PMC3567964 DOI: 10.1186/1742-2094-10-16] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 01/18/2013] [Indexed: 01/12/2023] Open
Abstract
Background Brain-derived neurotrophic factor (BDNF) has been shown to control microglial responses in neuropathic pain. Since adenosine A2A receptors (A2ARs) control neuroinflammation, as well as the production and function of BDNF, we tested to see if A2AR controls the microglia-dependent secretion of BDNF and the proliferation of microglial cells, a crucial event in neuroinflammation. Methods Murine N9 microglial cells were challenged with lipopolysaccharide (LPS, 100 ng/mL) in the absence or in the presence of the A2AR antagonist, SCH58261 (50 nM), as well as other modulators of A2AR signaling. The BDNF cellular content and secretion were quantified by Western blotting and ELISA, A2AR density was probed by Western blotting and immunocytochemistry and cell proliferation was assessed by BrdU incorporation. Additionally, the A2AR modulation of LPS-driven cell proliferation was also tested in primary cultures of mouse microglia. Results LPS induced time-dependent changes of the intra- and extracellular levels of BDNF and increased microglial proliferation. The maximal LPS-induced BDNF release was time-coincident with an LPS-induced increase of the A2AR density. Notably, removing endogenous extracellular adenosine or blocking A2AR prevented the LPS-mediated increase of both BDNF secretion and proliferation, as well as exogenous BDNF-induced proliferation. Conclusions We conclude that A2AR activation plays a mandatory role controlling the release of BDNF from activated microglia, as well as the autocrine/paracrine proliferative role of BDNF.
Collapse
Affiliation(s)
- Catarina Gomes
- Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra 3004-517, Portugal.
| | | | | | | | | | | | | |
Collapse
|
45
|
Song Y, Zeng Z, Jin C, Zhang J, Ding B, Zhang F. Protective effect of ginkgolide B against acute spinal cord injury in rats and its correlation with the Jak/STAT signaling pathway. Neurochem Res 2012; 38:610-9. [PMID: 23274522 DOI: 10.1007/s11064-012-0959-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/20/2012] [Accepted: 12/19/2012] [Indexed: 11/25/2022]
Abstract
This study aimed to investigate the correlation between ginkgolide B (GB) and the JAK/STAT signaling pathway and to explore its regulating effect on secondary cell apoptosis following spinal cord injury (SCI), to elucidate the protective mechanism GB against acute SCI. Sprague-Dawley rats were randomly divided into a sham-operated group, an SCI group, an SCI + GB group, an SCI + methylprednisolone (MP) group, and an SCI + specific JAK inhibitor AG490 group. A rat model of acute SCI was established using the modified Allen's method. At 4 h, 12 h, 1 day, 3 days, 7 days and 14 days after injury, injured T10 spinal cord specimens were harvested. GB significantly increased inclined plane test scores and Basso, Beattie, and Bresnahan scale scores in SCI rats from postoperative day 3 to day 14. The effect was equal to that of the positive control drug, MP. Western blot analysis showed that JAK(2) was significantly phosphorylated from 4 h after SCI, peaked at 12 h and gradually decreased thereafter, accompanied by phosphorylation of STAT(3) with a similar time course. GB was shown to significantly inhibit the phosphorylation of JAK(2) and STAT(3) in rats with SCI. It significantly increased the ratio of B cell CLL/lymphoma-2 (Bcl-2)/Bcl-2-associated X protein (Bax) protein expression at 24 h, led to an obvious down-regulation of caspase-3 gene and protein expression at 3 days, and significantly decreased the cell apoptosis index at each time point after SCI. This effect was similar to that obtained with the JAK-specific inhibitor, AG490. Our experimental findings indicated that GB can protect rats against acute SCI, and that its underlying mechanism may be related to the inhibition of JAK/STAT signaling pathway activation, improvement of the Bcl-2/Bax ratio, decreased caspase-3 gene and protein expression and further inhibition of secondary cell apoptosis following SCI.
Collapse
Affiliation(s)
- Yongxing Song
- Department of Orthopaedics, Hospital of Zhejiang General Corps of Armed Police Forces, Jiaxing, 314000, Zhejiang Province, People's Republic of China
| | | | | | | | | | | |
Collapse
|
46
|
A₁ adenosine receptor modulation of chemically and electrically evoked lumbar locomotor network activity in isolated newborn rat spinal cords. Neuroscience 2012; 222:191-204. [PMID: 22824428 DOI: 10.1016/j.neuroscience.2012.07.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Revised: 06/15/2012] [Accepted: 07/12/2012] [Indexed: 01/24/2023]
Abstract
It is not well-studied how the ubiquitous neuromodulator adenosine (ADO) affects mammalian locomotor network activities. We analyzed this here with focus on roles of 8-cyclopentyl-1,3-dipropylxanthine (DPCPX)-sensitive A(1)-type ADO receptors. For this, we recorded field potentials from ventral lumbar nerve roots and electrically stimulated dorsal roots in isolated newborn rat spinal cords. At ≥ 25μM, bath-applied ADO slowed synchronous bursting upon blockade of anion-channel-mediated synaptic inhibition by bicuculline (20 μM) plus strychnine (1 μM) and this depression was countered by DPCPX (1 μM) as tested at 100 μM ADO. ADO abolished this disinhibited rhythm at ≥ 500 μM. Contrary, the single electrical pulse-evoked dorsal root reflex, which was enhanced in bicuculline/strychnine-containing solution, persisted at all ADO doses (5 μM-2 mM). In control solution, ≥ 500 μM ADO depressed this reflex and pulse train-evoked bouts of alternating fictive locomotion; this inhibition was reversed by 1 μM DPCPX. ADO (5 μM-2 mM) did not depress, but stabilize alternating fictive locomotion evoked by serotonin (10 μM) plus N-methyl-d-aspartate (4-5 μM). Addition of DPCPX (1μM) to control solution did not change either the dorsal root reflex or rhythmic activities indicating lack of endogenous A(1) receptor activity. Our findings show A(1) receptor involvement in ADO depression of the dorsal root reflex, electrically evoked fictive locomotion and spontaneous disinhibited lumbar motor bursting. Contrary, chemically evoked fictive locomotion and the enhanced dorsal root reflex in disinhibited lumbar locomotor networks are resistant to ADO. Because ADO effects in standard solution occurred at doses that are notably higher than those occurring in vivo, we hypothesize that newborn rat locomotor networks are rather insensitive to this neuromodulator.
Collapse
|
47
|
Specific inhibition of the JNK pathway promotes locomotor recovery and neuroprotection after mouse spinal cord injury. Neurobiol Dis 2012; 46:710-21. [DOI: 10.1016/j.nbd.2012.03.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/31/2012] [Accepted: 03/01/2012] [Indexed: 12/23/2022] Open
|
48
|
Yu J, Huang X, Wu Q, Wang J, Yu X, Zhao P. Effect of A(2A) receptor antagonist (SCH 442416) on the mRNA expression of glutamate aspartate transporter and glutamine synthetase in rat retinal Müller cells under hypoxic conditions in vitro. Exp Ther Med 2012; 3:803-806. [PMID: 22969972 DOI: 10.3892/etm.2012.486] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Accepted: 01/30/2012] [Indexed: 11/06/2022] Open
Abstract
The purpose of the present study was to investigate the effect of the A(2A) receptor antagonist (SCH 442416) on the mRNA expression of glutamate aspartate transporter (GLAST) and glutamine synthetase (GS) in rat retinal Müller cells under hypoxic conditions in vitro. Immunofluorescent staining of GS and GFAP was used for the identification of Müller cells. The GLAST and GS mRNA expression of Müller cells treated with 0.1, 1 and 10 μM SCH 442416 under hypoxic conditions was examined by real-time PCR. Müller cells increased the mRNA expression of GLAST under hypoxic conditions; those treated with 0.1 μM SCH 442416 showed a further significant increase in the mRNA expression of GLAST in vitro. Although the mRNA expression of GS was decreased under hypoxic conditions, the mRNA expression was increased when Müller cells were treated with 0.1 μM SCH 442416. A(2A) receptor antagonist increased the GLAST and GS expression of Müller cells and accelerated the clearance of extracellular glutamate under hypoxic conditions in vitro.
Collapse
Affiliation(s)
- Jun Yu
- Department of Ophthalmology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | | | | | | | | | | |
Collapse
|
49
|
Rico EP, Rosemberg DB, Seibt KJ, Capiotti KM, Da Silva RS, Bonan CD. Zebrafish neurotransmitter systems as potential pharmacological and toxicological targets. Neurotoxicol Teratol 2011; 33:608-17. [PMID: 21907791 DOI: 10.1016/j.ntt.2011.07.007] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/20/2011] [Accepted: 07/20/2011] [Indexed: 01/23/2023]
Abstract
Recent advances in neurobiology have emphasized the study of brain structure and function and its association with numerous pathological and toxicological events. Neurotransmitters are substances that relay, amplify, and modulate electrical signals between neurons and other cells. Neurotransmitter signaling mediates rapid intercellular communication by interacting with cell surface receptors, activating second messenger systems and regulating the activity of ion channels. Changes in the functional balance of neurotransmitters have been implicated in the failure of central nervous system function. In addition, abnormalities in neurotransmitter production or functioning can be induced by several toxicological compounds, many of which are found in the environment. The zebrafish has been increasingly used as an animal model for biomedical research, primarily due to its genetic tractability and ease of maintenance. These features make this species a versatile tool for pre-clinical drug discovery and toxicological investigations. Here, we present a review regarding the role of different excitatory and inhibitory neurotransmitter systems in zebrafish, such as dopaminergic, serotoninergic, cholinergic, purinergic, histaminergic, nitrergic, glutamatergic, glycinergic, and GABAergic systems, and emphasizing their features as pharmacological and toxicological targets. The increase in the global knowledge of neurotransmitter systems in zebrafish and the elucidation of their pharmacological and toxicological aspects may lead to new strategies and appropriate research priorities to offer insights for biomedical and environmental research.
Collapse
Affiliation(s)
- E P Rico
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600-Anexo, Porto Alegre, RS, Brazil
| | | | | | | | | | | |
Collapse
|