1
|
Zhang S, Gao Z, Feng L, Li M. Prevention and Treatment Strategies for Alzheimer's Disease: Focusing on Microglia and Astrocytes in Neuroinflammation. J Inflamm Res 2024; 17:7235-7259. [PMID: 39421566 PMCID: PMC11484773 DOI: 10.2147/jir.s483412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease characterized by its insidious onset and progressive development, making it the most common form of dementia. Despite its prevalence, the exact causes and mechanisms responsible for AD remain unclear. Recent studies have highlighted that inflammation in the central nervous system (CNS) plays a crucial role in both the initiation and progression of AD. Neuroinflammation, an immune response within the CNS triggered by glial cells in response to various stimuli, such as nerve injury, infection, toxins, or autoimmune reactions, has emerged as a significant factor alongside amyloid deposition and neurofibrillary tangles (NFTs) commonly associated with AD. This article aims to provide an overview of the most recent research regarding the involvement of neuroinflammation in AD, with a particular focus on elucidating the specific mechanisms involving microglia and astrocytes. By exploring these intricate processes, a new theoretical framework can be established to further probe the impact of neuroinflammation on the development and progression of AD. Through a deeper understanding of these underlying mechanisms, potential targets for therapeutic interventions and novel treatment strategies can be identified in the ongoing battle against AD.
Collapse
Affiliation(s)
- Shenghao Zhang
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Zhejianyi Gao
- Department of Orthopaedics, Fushun Hospital of Chinese Medicine, Fushun, Liaoning Province, 113008, People’s Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, 271000, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| |
Collapse
|
2
|
Li L, Li W, Jiang W, Xu R. Sulbactam protects neurons against double neurotoxicity of amyloid beta and glutamate load by upregulating glial glutamate transporter 1. Cell Death Discov 2024; 10:64. [PMID: 38320997 PMCID: PMC10847450 DOI: 10.1038/s41420-024-01827-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/08/2024] Open
Abstract
Amyloid beta (Abeta) synergistically enhances excitotoxicity of glutamate load by impairing glutamate transporter 1 (GLT1) expression and function, which exacerbates the development of Alzheimer's disease (AD). Our previous studies suggested that sulbactam can upregulate the expression levels and capacity of GLT1. Therefore, this study aims to investigate whether sulbactam improves neuronal tolerance against neurotoxicity of Abeta and glutamate load by up-regulating GLT1 in primary neuron-astrocyte co-cultures. Early postnatal P0-P1 Wistar rat pups' cortices were collected for primary neuron-astrocyte cultures. Hoechst-propidium iodide (HO-PI) stain and lactate dehydrogenase (LDH) assays were used to analyze neuronal death. Cell counting kit 8 (CCK8) was applied to determine cell viability. Immunofluorescence staining and western blotting were used to assess protein expressions including GLT1, B-cell lymphoma 2 (BCL2), BCL2 associated X (BAX), and cleaved caspase 3 (CCP3). Under the double effect of Abeta and glutamate load, more neurons were lost than that induced by Abeta or glutamate alone, shown as decreased cell viability, increased LDH concentration in the cultural medium, HO-PI positive stains, high CCP3 expression, and high BAX/BCL2 ratio resulting from increased BAX and decreased BCL2 expressions. Notably, pre-incubation with sulbactam significantly attenuated the neuronal loss and activation of apoptosis induced by both Abeta and glutamate in a dose-dependent manner. Simultaneously, both astrocytic and neuronal GLT1 expressions were upregulated after sulbactam incubation. Taken together, it could be concluded that sulbactam protected neurons against double neurotoxicity of Abeta and glutamate load by upregulating GLT1 expression. The conclusion provides evidence for potential intervention using sulbactam in AD research.
Collapse
Affiliation(s)
- Li Li
- The Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Wenbin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wei Jiang
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Vascular Homeostasis Key Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Renhao Xu
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Vascular Homeostasis Key Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
3
|
Rao GN, Jupudi S, Justin A. A Review on Neuroinflammatory Pathway Mediating Through Ang-II/AT1 Receptors and a Novel Approach for the Treatment of Cerebral Ischemia in Combination with ARB's and Ceftriaxone. Ann Neurosci 2024; 31:53-62. [PMID: 38584983 PMCID: PMC10996871 DOI: 10.1177/09727531231182554] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/16/2023] [Indexed: 04/09/2024] Open
Abstract
Background Ischemic stroke is one of the prevalent neurodegenerative disorders; it is generally characterized by sudden abruption of blood flow due to thromboembolism and vascular abnormalities, eventually impairing the supply of oxygen and nutrients to the brain for its metabolic needs. Oxygen-glucose deprived conditions provoke the release of excessive glutamate, which causes excitotoxicity. Summary Recent studies suggest that circulatory angiotensin-II (Ang-II) has an imperative role in initiating detrimental events through binding central angiotensin 1 (AT1) receptors. Insufficient energy metabolites and essential ions often lead to oxidative stress during ischemic reperfusion, which leads to the release of proinflammatory mediators such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and cytokines like interleukin-18 (IL-18) and interleukin- 1beta (IL-1β). The transmembrane glutamate transporters, excitatory amino acid transporter-2 (EAAT-2), which express in astroglial cells, have a crucial role in the clearance of glutamate from its releasing site and convert glutamate into glutamine in normal circumstances of brain physiology. Key Message During cerebral ischemia, an impairment or dysfunction of EAAT-2 attributes the risk of delayed neuronal cell death. Earlier studies evidencing that angiotensin receptor blockers (ARB) attenuate neuroinflammation by inhibiting the Ang-II/AT1 receptor-mediated inflammatory pathway and that ceftriaxone ameliorates the excitotoxicity-induced neuronal deterioration by enhancing the transcription and expression of EAAT-2 via the nuclear transcriptional factor kappa-B (NF-kB) signaling pathway. The present review will briefly discuss the mechanisms involved in Ang-II/AT1-mediated neuroinflammation, ceftriaxone-induced EAAT-2 expression, and the repurposing hypothesis of the novel combination of ARBs and ceftriaxone for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Gaddam Narasimha Rao
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Srikanth Jupudi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Antony Justin
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| |
Collapse
|
4
|
Liu LZ, Fan SJ, Gao JX, Li WB, Xian XH. Ceftriaxone ameliorates hippocampal synapse loss by inhibiting microglial/macrophages activation in glial glutamate transporter-1 dependent manner in the APP/PS1 mouse model of Alzheimer's disease. Brain Res Bull 2023:110683. [PMID: 37301482 DOI: 10.1016/j.brainresbull.2023.110683] [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] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023]
Abstract
Synapse loss is a major contributor to cognitive dysfunction in Alzheimer's disease (AD). Impairments in the expression and/or glutamate uptake activity of glia glutamate transporter-1 (GLT-1) contribute to synapse loss in AD. Hence, targeting the restoration of GLT-1 activity may have potential for alleviating synapse loss in AD. Ceftriaxone (Cef) can upregulate the expression and glutamate uptake activity of GLT-1 in many disease models, including those for AD. The present study investigated the effects of Cef on synapse loss and the role of GLT-1 using APP/PS1 transgenic and GLT-1 knockdown APP/PS1 AD mice. Furthermore, the involvement of microglia in the process was investigated due to its important role in synapse loss in AD. We found that Cef treatment significantly ameliorated synapse loss and dendritic degeneration in APP/PS1 AD mice, evidenced by an increased dendritic spine density, decreased dendritic beading density, and upregulated levels of postsynaptic density protein 95 (PSD95) and synaptophysin. The effects of Cef were suppressed by GLT-1 knockdown in GLT-1+/-/APP/PS1 AD mice. Simultaneously, Cef treatment inhibited ionized calcium binding adapter molecule 1 (Iba1) expression, decreased the proportion of CD11b+CD45hi cells, declined interleukin-6 (IL-6) content, and reduced the co-expression of Iba1 with PSD95 or synaptophysin in APP/PS1 AD mice. In conclusion, Cef treatment ameliorated synapse loss and dendritic degeneration in APP/PS1 AD mice in a GLT-1-dependent manner, and the inhibitory effect of Cef on the activation of microglia/macrophages and their phagocytosis for synaptic elements contributed to the mechanism.
Collapse
Affiliation(s)
- Li-Zhe Liu
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, P.R. China; Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, P. R. China.
| | - Shu-Juan Fan
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, P.R. China; Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, P. R. China.
| | - Jun-Xia Gao
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, P.R. China; Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, P. R. China.
| | - Wen-Bin Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, P.R. China; Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, P. R. China.
| | - Xiao-Hui Xian
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, P.R. China; Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, P. R. China.
| |
Collapse
|
5
|
Sattayakhom A, Kalarat K, Rakmak T, Tapechum S, Monteil A, Punsawad C, Palipoch S, Koomhin P. Effects of Ceftriaxone on Oxidative Stress and Inflammation in a Rat Model of Chronic Cerebral Hypoperfusion. Behav Sci (Basel) 2022; 12:bs12080287. [PMID: 36004858 PMCID: PMC9404883 DOI: 10.3390/bs12080287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Ceftriaxone (CTX) exerts a neuroprotective effect by decreasing glutamate excitotoxicity. We further studied the underlying mechanisms and effects of CTX early post-treatment on behavior in a cerebral hypoperfusion rats. The rats’ common carotid arteries (2VO) were permanently ligated. CTX was treated after ischemia. Biochemical studies were performed to assess antioxidative stress and inflammation. Behavioral and histological studies were then tested on the ninth week after vessel ligation. The 2VO rats showed learning and memory deficits as well as working memory impairments without any motor weakness. The treatment with CTX was found to attenuate white matter damage, MDA production, and interleukin 1 beta and tumor necrosis factor alpha production, mainly in the hippocampal area. Moreover, CTX treatment could increase the expression of glia and the glial glutamate transporters, and the neuronal glutamate transporter. Taken together, our data indicate the neuroprotective mechanisms of CTX involving the upregulation of glutamate transporters’ expression. This increased expression contributes to a reduction in glutamate excitotoxicity and oxidative stress as well as pro-inflammatory cytokine production, thus resulting in the protection of neurons and tissue from further damage. The present study highlights the mechanism of the effect of CTX treatment and of the underlying ischemia-induced neuronal damage.
Collapse
Affiliation(s)
- Apsorn Sattayakhom
- School of Allied Health Sciences, Walailak University, Nakhonsithammarat 80160, Thailand
- Center of Excellence in Innovation on Essential Oil, Walailak University, Nakhonsithammarat 80160, Thailand
| | - Kosin Kalarat
- School of Informatics, Walailak University, Nakhonsithammarat 80160, Thailand
| | - Thatdao Rakmak
- School of Liberal Arts, Walailak University, Nakhonsithammarat 80160, Thailand
| | - Sompol Tapechum
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Arnaud Monteil
- Institutde Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Chuchard Punsawad
- School of Medicine, Walailak University, Nakhonsithammarat 80160, Thailand
| | - Sarawoot Palipoch
- School of Medicine, Walailak University, Nakhonsithammarat 80160, Thailand
| | - Phanit Koomhin
- Center of Excellence in Innovation on Essential Oil, Walailak University, Nakhonsithammarat 80160, Thailand
- School of Medicine, Walailak University, Nakhonsithammarat 80160, Thailand
- Correspondence: ; Tel.: +66-(0)-95-295-0550
| |
Collapse
|
6
|
Abulseoud OA, Alasmari F, Hussein AM, Sari Y. Ceftriaxone as a Novel Therapeutic Agent for Hyperglutamatergic States: Bridging the Gap Between Preclinical Results and Clinical Translation. Front Neurosci 2022; 16:841036. [PMID: 35864981 PMCID: PMC9294323 DOI: 10.3389/fnins.2022.841036] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulation of glutamate homeostasis is a well-established core feature of neuropsychiatric disorders. Extracellular glutamate concentration is regulated by glutamate transporter 1 (GLT-1). The discovery of a beta-lactam antibiotic, ceftriaxone (CEF), as a safe compound with unique ability to upregulate GLT-1 sparked the interest in testing its efficacy as a novel therapeutic agent in animal models of neuropsychiatric disorders with hyperglutamatergic states. Indeed, more than 100 preclinical studies have shown the efficacy of CEF in attenuating the behavioral manifestations of various hyperglutamatergic brain disorders such as ischemic stroke, amyotrophic lateral sclerosis (ALS), seizure, Huntington’s disease, and various aspects of drug use disorders. However, despite rich and promising preclinical data, only one large-scale clinical trial testing the efficacy of CEF in patients with ALS is reported. Unfortunately, in that study, there was no significant difference in survival between placebo- and CEF-treated patients. In this review, we discussed the translational potential of preclinical efficacy of CEF based on four different parameters: (1) initiation of CEF treatment in relation to induction of the hyperglutamatergic state, (2) onset of response in preclinical models in relation to onset of GLT-1 upregulation, (3) mechanisms of action of CEF on GLT-1 expression and function, and (4) non-GLT-1-mediated mechanisms for CEF. Our detailed review of the literature brings new insights into underlying molecular mechanisms correlating the preclinical efficacy of CEF. We concluded here that CEF may be clinically effective in selected cases in acute and transient hyperglutamatergic states such as early drug withdrawal conditions.
Collapse
Affiliation(s)
- Osama A. Abulseoud
- Department of Psychiatry and Psychology, Alex School of Medicine at Mayo Clinic, Phoenix, AZ, United States
- *Correspondence: Osama A. Abulseoud,
| | - Fawaz Alasmari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
- Youssef Sari,
| |
Collapse
|
7
|
Gagnon DJ, Ryzhov SV, May MA, Riker RR, Geller B, May TL, Bockian S, deKay JT, Eldridge A, Van der Kloot T, Lerwick P, Lord C, Lucas FL, Mailloux P, McCrum B, Searight M, Wirth J, Zuckerman J, Sawyer D, Seder DB. Ceftriaxone to PRevent pneumOnia and inflammaTion aftEr Cardiac arresT (PROTECT): study protocol for a randomized, placebo-controlled trial. Trials 2022; 23:197. [PMID: 35246202 PMCID: PMC8895836 DOI: 10.1186/s13063-022-06127-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/23/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Pneumonia is the most common infection after out-of-hospital cardiac arrest (OHCA) occurring in up to 65% of patients who remain comatose after return of spontaneous circulation. Preventing infection after OHCA may (1) reduce exposure to broad-spectrum antibiotics, (2) prevent hemodynamic derangements due to local and systemic inflammation, and (3) prevent infection-associated morbidity and mortality. METHODS The ceftriaxone to PRevent pneumOnia and inflammaTion aftEr Cardiac arrest (PROTECT) trial is a randomized, placebo-controlled, single-center, quadruple-blind (patient, treatment team, research team, outcome assessors), non-commercial, superiority trial to be conducted at Maine Medical Center in Portland, Maine, USA. Ceftriaxone 2 g intravenously every 12 h for 3 days will be compared with matching placebo. The primary efficacy outcome is incidence of early-onset pneumonia occurring < 4 days after mechanical ventilation initiation. Concurrently, T cell-mediated inflammation bacterial resistomes will be examined. Safety outcomes include incidence of type-one immediate-type hypersensitivity reactions, gallbladder injury, and Clostridioides difficile-associated diarrhea. The trial will enroll 120 subjects over approximately 3 to 4 years. DISCUSSION The PROTECT trial is novel in its (1) inclusion of OHCA survivors regardless of initial heart rhythm, (2) use of a low-risk antibiotic available in the USA that has not previously been tested after OHCA, (3) inclusion of anti-inflammatory effects of ceftriaxone as a novel mechanism for improved clinical outcomes, and (4) complete metagenomic assessment of bacterial resistomes pre- and post-ceftriaxone prophylaxis. The long-term goal is to develop a definitive phase III trial powered for mortality or functional outcome. TRIAL REGISTRATION ClinicalTrials.gov NCT04999592 . Registered on August 10, 2021.
Collapse
Affiliation(s)
- David J Gagnon
- Department of Pharmacy, Maine Medical Center, Portland, ME, USA.
- Maine Medical Center Research Institute, Scarborough, ME, USA.
- Tufts University School of Medicine, Boston, MA, USA.
| | - Sergey V Ryzhov
- Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Meghan A May
- University of New England College of Osteopathic Medicine, Biddeford, ME, USA
| | - Richard R Riker
- Tufts University School of Medicine, Boston, MA, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Bram Geller
- Tufts University School of Medicine, Boston, MA, USA
- Maine Medical Partners, MaineHealth Cardiology, Scarborough, ME, USA
| | - Teresa L May
- Maine Medical Center Research Institute, Scarborough, ME, USA
- Tufts University School of Medicine, Boston, MA, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Sarah Bockian
- Maine Medical Center Neuroscience Institute, Maine Medical Center, Portland, ME, USA
| | - Joanne T deKay
- Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Ashley Eldridge
- Maine Medical Center Neuroscience Institute, Maine Medical Center, Portland, ME, USA
| | | | - Patricia Lerwick
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Christine Lord
- Maine Medical Center Neuroscience Institute, Maine Medical Center, Portland, ME, USA
| | - F Lee Lucas
- Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Patrick Mailloux
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Barbara McCrum
- Maine Medical Center Neuroscience Institute, Maine Medical Center, Portland, ME, USA
| | - Meghan Searight
- Maine Medical Center Neuroscience Institute, Maine Medical Center, Portland, ME, USA
| | - Joel Wirth
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | | | - Douglas Sawyer
- Maine Medical Center Research Institute, Scarborough, ME, USA
- Maine Medical Partners, MaineHealth Cardiology, Scarborough, ME, USA
| | - David B Seder
- Maine Medical Center Research Institute, Scarborough, ME, USA
- Tufts University School of Medicine, Boston, MA, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| |
Collapse
|
8
|
Sulbactam improves binding property and uptake capacity of glutamate transporter-1 and decreases glutamate concentration in the CA1 region of hippocampus of global brain ischemic rats. Amino Acids 2021; 53:1649-1661. [PMID: 34716803 DOI: 10.1007/s00726-021-03088-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/07/2021] [Indexed: 10/19/2022]
Abstract
Glutamate transporter-1 (GLT-1) removes most glutamate in the synaptic cleft. Sulbactam confers neuronal protection against ischemic insults in the hippocampal CA1 region accompanied by the upregulation of GLT-1 expression in rats. The present study further investigates the effect of sulbactam on the binding property and uptake capacity of GLT-1 for glutamate, and the change in extracellular glutamate concentration in the hippocampal CA1 region of rats with global brain ischemia. The binding property and uptake capacity of GLT-1 were measured using a radioligand binding and uptake assay, respectively, with L-3H-glutamate. The extracellular glutamate concentration was detected using microdialysis and high-performance liquid chromatography-mass spectrometry. Neuropathological evaluation was performed based on thionin staining. It was shown that sulbactam pre-treatment changed GLT-1 binding property, including increased Bmax and decreased Kd values, increased GLT-1 uptake capacity for glutamate, and inhibited the elevation of extracellular glutamate concentration in rats with global cerebral ischemia. These effects of sulbactam were accompanied by its neuronal protection on the hippocampal CA1 neurons against delayed neuronal death resulted from ischemic insult. Furthermore, administration of GLT-1 antisense oligodeoxynucleotides, which inhibited the expression of GLT-1, blocked the aforementioned sulbactam-related effects, which suggested that GLT-1 upregulation mediated the above effect although other mechanisms independent of the upregulation of GLT-1 expression could not be excluded. It could be concluded that sulbactam improves the binding property and uptake capacity of GLT-1 for glutamate and then reduces the glutamate concentration and excitotoxicity during global cerebral ischemia, which contributes to the neuroprotection of sulbactam against brain ischemia.
Collapse
|
9
|
Ceftriaxone Treatment Weakens Long-Term Synaptic Potentiation in the Hippocampus of Young Rats. Int J Mol Sci 2021; 22:ijms22168417. [PMID: 34445137 PMCID: PMC8395093 DOI: 10.3390/ijms22168417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/02/2022] Open
Abstract
Disrupted glutamate clearance in the synaptic cleft leads to synaptic dysfunction and neurological diseases. Decreased glutamate removal from the synaptic cleft is known to cause excitotoxicity. Data on the physiological effects of increased glutamate clearance are contradictory. This study investigated the consequences of ceftriaxone (CTX), an enhancer of glutamate transporter 1 expression, treatment on long-term synaptic potentiation (LTP) in the hippocampus of young rats. In this study, 5-day administration of CTX (200 mg/kg) significantly weakened LTP in CA3-CA1 synapses. As shown by electrophysiological recordings, LTP attenuation was associated with weakening of N-Methyl-D-aspartate receptor (NMDAR)-dependent signaling in synapses. However, PCR analysis did not show downregulation of NMDAR subunits or changes in the expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits. We assume that extracellular burst stimulation activates fewer synapses in CTX-treated animals because increased glutamate reuptake results in reduced spillover, and neighboring synapses do not participate in neurotransmission. Attenuation of LTP was not accompanied by noticeable behavioral changes in the CTX group, with no behavioral abnormalities observed in the open field test or Morris water maze test. Thus, our experiments show that increased glutamate clearance can impair long-term synaptic plasticity and that this phenomenon can be considered a potential side effect of CTX treatment.
Collapse
|
10
|
Sun YW, Zhang LY, Gong SJ, Hu YY, Zhang JG, Xian XH, Li WB, Zhang M. The p38 MAPK/NF-κB pathway mediates GLT-1 up-regulation during cerebral ischemic preconditioning-induced brain ischemic tolerance in rats. Brain Res Bull 2021; 175:224-233. [PMID: 34343641 DOI: 10.1016/j.brainresbull.2021.07.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Our previous finding suggests that p38 MAPK contributes to the GLT-1 upregulation during induction of brain ischemic tolerance by cerebral ischemic preconditioning (CIP), however, the underlying mechanism is still unclear. Here, we investigated the molecular mechanisms underlying the CIP-induced GLT-1 upregulation by using Western blotting, Co-immunoprecipitation (Co-IP), electrophoretic mobility shift assay (EMSA) and thionin staining in rat hippocampus CA1 subset. We found that application of BAY11-7082 (an inhibitor of NF-κB), or dihydrokainate (an inhibitor of GLT-1), or SB203580 (an inhibitor of p38 MAPK) could attenuate the CIP-induced neuronal protection in hippocampus CA1 region of rats. Moreover, CIP caused rapid activation of NF-κB, as evidenced by nuclear translocation of NF-κB p50 protein, which led to active p50/p65 dimer formation and increased DNA binding activity. GLT-1 was also increased after CIP. Pretreatment with BAY11-7082 blocked the CIP-induced GLT-1 upregulation. The above results suggest that NF-κB participates in GLT-1 up-regulation during the induction of brain ischemic tolerance by CIP. We also found that pretreatment with SB203580 caused significant reduction of NF-κB p50 protein in nucleus, NF-κB p50/p65 dimer nuclear translocation and DNA binding activity of NF-κB. Together, we conclude that p38 MAPK/NF-κB pathway participates in the mediation of GLT-1 up-regulation during the induction of brain ischemic tolerance induced by CIP.
Collapse
Affiliation(s)
- Ya-Wei Sun
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Xing Tai People's Hospital, 16 Hong Xing Road, Xing Tai, 054001, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Shu-Juan Gong
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, People's Republic of China.
| |
Collapse
|
11
|
Li HH, Lin PJ, Wang WH, Tseng LH, Tung H, Liu WY, Lin CL, Liu CH, Liao WC, Hung CS, Ho YJ. Treatment effects of the combination of ceftriaxone and valproic acid on neuronal and behavioural functions in a rat model of epilepsy. Exp Physiol 2021; 106:1814-1828. [PMID: 34086374 DOI: 10.1113/ep089624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/03/2021] [Indexed: 01/29/2023]
Abstract
NEW FINDINGS What is the central question of this study? Imbalance of activities between GABAergic and glutamatergic systems is involved in epilepsy. It is not known whether simultaneously increasing GABAergic and decreasing glutamatergic activity using valproic acid and ceftriaxone, respectively, leads to better seizure control. What is the central question of this study? Ceftriaxone suppressed seizure and cognitive deficits and restored neuronal density and the number of newborn cells in the hippocampus in a rat model of epilepsy. Combined treatment with ceftriaxone and valproic acid showed additive effects in seizure suppression. ABSTRACT The pathophysiology of epilepsy is typically considered as an imbalance between inhibitory GABA and excitatory glutamate neurotransmission. Valproic acid (Val), a GABA agonist, is one of the first-line antiepileptic drugs in the treatment of epilepsy, but it exhibits adverse effects. Ceftriaxone (CEF) elevates expression of glutamate transporter-1, enhances the reuptake of synaptic glutamate, increases the number of newborn cells and exhibits neuroprotective effects in animal studies. In this study, we evaluated effects of the combination of CEF and Val on behavioural and neuronal measures in a rat epilepsy model. Male Wistar rats were injected i.p. with pentylenetetrazol (35 mg/kg, every other day for 13 days) to induce the epilepsy model. Ceftriaxone (10 or 50 mg/kg), Val (50 or 100 mg/kg) or the combination of CEF and Val were injected daily after the fourth pentylenetetrazol injection for seven consecutive days. Epileptic rats exhibited seizure and impairments in motor and cognitive functions. Treatment with CEF and Val reduced the seizure and enhanced motor and cognitive functions in a dose-dependent manner. The combination of CEF (10 mg/kg) and Val (50 mg/kg) improved behaviours considerably. Histologically, compared with control animals, epileptic rats exhibited lower neuronal density and a reduction in hippocampal newborn cells but higher apoptosis in the basolateral amygdala, all of which were restored by the treatment with CEF, Val or the combination of CEF and Val. The study findings demonstrated that the combination of low doses of CEF and Val has beneficial effects on seizure suppression, neuroprotection and improvement in motor and cognitive functions in epilepsy.
Collapse
Affiliation(s)
- Hsin-Hua Li
- Department of Medical Research, Institute of Medicine, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Pin-Jiun Lin
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Wei-Han Wang
- Department of Psychology, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Li-Ho Tseng
- Graduate School of Environmental Management, Tajen University, Pingtung, Taiwan, Republic of China
| | - Hsin Tung
- Division of Epilepsy, Center of Faculty Development, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Wen-Yuan Liu
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Chih-Li Lin
- Department of Medical Research, Institute of Medicine, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Chiung-Hui Liu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Wen-Chieh Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Ching-Sui Hung
- Occupational Safety and Health Office, Taipei City Hospital, Taipei, Taiwan, Republic of China
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
12
|
Wilkie CM, Barron JC, Brymer KJ, Barnes JR, Nafar F, Parsons MP. The Effect of GLT-1 Upregulation on Extracellular Glutamate Dynamics. Front Cell Neurosci 2021; 15:661412. [PMID: 33841104 PMCID: PMC8032948 DOI: 10.3389/fncel.2021.661412] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022] Open
Abstract
Pharmacological upregulation of glutamate transporter-1 (GLT-1), commonly achieved using the beta-lactam antibiotic ceftriaxone, represents a promising therapeutic strategy to accelerate glutamate uptake and prevent excitotoxic damage in neurological conditions. While excitotoxicity is indeed implicated in numerous brain diseases, it is typically restricted to select vulnerable brain regions, particularly in early disease stages. In healthy brain tissue, the speed of glutamate uptake is not constant and rather varies in both an activity- and region-dependent manner. Despite the widespread use of ceftriaxone in disease models, very little is known about how such treatments impact functional measures of glutamate uptake in healthy tissue, and whether GLT-1 upregulation can mask the naturally occurring activity-dependent and regional heterogeneities in uptake. Here, we used two different compounds, ceftriaxone and LDN/OSU-0212320 (LDN), to upregulate GLT-1 in healthy wild-type mice. We then used real-time imaging of the glutamate biosensor iGluSnFR to investigate functional consequences of GLT-1 upregulation on activity- and regional-dependent variations in glutamate uptake capacity. We found that while both ceftriaxone and LDN increased GLT-1 expression in multiple brain regions, they did not prevent activity-dependent slowing of glutamate clearance nor did they speed basal clearance rates, even in areas characterized by slow uptake (e.g., striatum). Unexpectedly, ceftriaxone but not LDN decreased glutamate release in the cortex, suggesting that ceftriaxone may alter release properties independent of its effects on GLT-1 expression. In sum, our data demonstrate the complexities of glutamate uptake by showing that GLT-1 expression does not necessarily translate to accelerated uptake. Furthermore, these data suggest that the mechanisms underlying activity- and regional-dependent differences in glutamate dynamics are independent of GLT-1 expression levels.
Collapse
Affiliation(s)
- Crystal M Wilkie
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Jessica C Barron
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Kyle J Brymer
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Jocelyn R Barnes
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Firoozeh Nafar
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| |
Collapse
|
13
|
Li LL, Ke XY, Jiang C, Qin SQ, Liu YY, Xian XH, Liu LZ, He JC, Chen YM, An HF, Sun N, Hu YH, Wang Y, Zhang LN, Lu QY. Na + , K + -ATPase participates in the protective mechanism of rat cerebral ischemia-reperfusion through the interaction with glutamate transporter-1. Fundam Clin Pharmacol 2021; 35:870-881. [PMID: 33481320 DOI: 10.1111/fcp.12652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/19/2021] [Indexed: 11/30/2022]
Abstract
Glutamate excitotoxicity in cerebral ischemia/reperfusion is an important cause of neurological damage. The aim of this study was to investigate the mechanism of Na+, K+-ATPase (NKA) involved in l ow concentration of ouabain (Oua, activating NKA)-induced protection of rat cerebral ischemia-reperfusion injury. The 2,3,5-triphenyltetrazolium chloride (TTC) staining and neurological deficit scores (NDS) were performed to evaluate rat cerebral injury degree respectively at 2 h, 6 h, 1 d and 3 d after reperfusion of middle cerebral artery occlusion (MCAO) 2 h in rats. NKA α1/α2 subunits and glutamate transporter-1 (GLT-1) protein expression were investigated by Western blotting. The cerebral infarct volume ratio were evidently decreased in Oua group vs MCAO/R group at 1 d and 3 d after reperfusion of 2 h MCAO in rats (*p < 0.05 ). Moreover, NDS were not significantly different (p > 0.05 ). NKA α1 was decreased at 6 h and 1 d after reperfusion of 2 h MCAO in rats, and was improved in Oua group. However, NKA α1 and α2 were increased at 3 d after reperfusion of 2 h MCAO in rats, and was decreased in Oua group. GLT-1 was decreased at 6 h, 1 d and 3 d after reperfusion of 2 h MCAO in rats, and was improved in Oua group. These data indicated that l ow concentration of Oua could improve MCAO/R injury through probably changing NKA α1/α2 and GLT-1 protein expression, then increasing GLT-1 function and promoting Glu transport and absorption, which could be useful to determine potential therapeutic strategies for patients with stroke. Low concentration of Oua improved rat MCAO/R injury via NKA α1/α2 and GLT-1.
Collapse
Affiliation(s)
- Lin-Lin Li
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Xue-Ying Ke
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Chen Jiang
- Forensic Medical College, Hebei Medical University, Hebei, China
| | - Shi-Qi Qin
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Yang-Yang Liu
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, Hebei, China
| | - Li-Zhe Liu
- Department of Pathophysiology, Hebei Medical University, Hebei, China
| | - Jin-Chen He
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Ya-Meng Chen
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Hong-Fei An
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Nan Sun
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Yue-Hua Hu
- Basic Medical College, Hebei Medical University, Hebei, China
| | - Yan Wang
- North China University of Science and Technology Affiliated Hospital, Hebei, China
| | - Li-Nan Zhang
- Department of Pathophysiology, Hebei Medical University, Hebei, China
| | - Qi-Yong Lu
- Department of Neurosurgery, Hengshui Fifth People's Hospital, Hebei, China
| |
Collapse
|
14
|
Tejeda-Bayron FA, Rivera-Aponte DE, Malpica-Nieves CJ, Maldonado-Martínez G, Maldonado HM, Skatchkov SN, Eaton MJ. Activation of Glutamate Transporter-1 (GLT-1) Confers Sex-Dependent Neuroprotection in Brain Ischemia. Brain Sci 2021; 11:76. [PMID: 33429955 PMCID: PMC7827447 DOI: 10.3390/brainsci11010076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/22/2022] Open
Abstract
Stroke is one of the leading causes of long-term disability. During ischemic stroke, glutamate is released, reuptake processes are impaired, and glutamate promotes excitotoxic neuronal death. Astrocytic glutamate transporter 1 (GLT-1) is the major transporter responsible for removing excess glutamate from the extracellular space. A translational activator of GLT-1, LDN/OSU 0212320 (LDN) has been previously developed with beneficial outcomes in epileptic animal models but has never been tested as a potential therapeutic for ischemic strokes. The present study evaluated the effects of LDN on stroke-associated brain injury. Male and female mice received LDN or vehicle 24 h before or 2 h after focal ischemia was induced in the sensorimotor cortex. Sensorimotor performance was determined using the Rung Ladder Walk and infarct area was assessed using triphenyltetrazolium chloride staining. Males treated with LDN exhibited upregulated GLT-1 protein levels, significantly smaller infarct size, and displayed better sensorimotor performance in comparison to those treated with vehicle only. In contrast, there was no upregulation of GLT-1 protein levels and no difference in infarct size or sensorimotor performance between vehicle- and LDN-treated females. Taken together, our results indicate that the GLT-1 translational activator LDN improved stroke outcomes in young adult male, but not female mice.
Collapse
Affiliation(s)
- Flavia A. Tejeda-Bayron
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (F.A.T.-B.); (D.E.R.-A.); (C.J.M.-N.)
| | - David E. Rivera-Aponte
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (F.A.T.-B.); (D.E.R.-A.); (C.J.M.-N.)
| | - Christian J. Malpica-Nieves
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (F.A.T.-B.); (D.E.R.-A.); (C.J.M.-N.)
| | - Gerónimo Maldonado-Martínez
- School of Chiropractic, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA;
- Biology Department, University of Puerto Rico—Río Piedras Campus, Río Piedras, PR 00924-2537, USA
| | - Héctor M. Maldonado
- Department of Pharmacology, School of Medicine, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA;
| | - Serguei N. Skatchkov
- Department of Physiology and Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA;
| | - Misty J. Eaton
- Department of Biochemistry, School of Medicine, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (F.A.T.-B.); (D.E.R.-A.); (C.J.M.-N.)
| |
Collapse
|
15
|
Rodolphi MS, Kopczynski A, Carteri RB, Sartor M, Fontella FU, Feldmann M, Hansel G, Strogulski NR, Portela LV. Glutamate transporter-1 link astrocytes with heightened aggressive behavior induced by steroid abuse in male CF1 mice. Horm Behav 2021; 127:104872. [PMID: 33069754 DOI: 10.1016/j.yhbeh.2020.104872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 11/21/2022]
Abstract
The astrocytic glutamate transporter GLT-1 performs glutamate uptake thereby mediating NMDAr responses in neurons. Ceftriaxone (CEF) upregulates astrocytic GLT-1 expression/activity, which could counteract excessive glutamate levels and aggressive behavior induced by anabolic synthetic steroids such as nandrolone decanoate (ND). Here, adult male CF-1 mice were allocated to oil (VEH), ND, CEF, and ND/CEF groups. Mice were subcutaneously (s.c.) injected with ND (15 mg/kg) or VEH for 19 days, and received intraperitoneal (i.p.) injections of CEF (200 mg/kg) or saline for 5 days. The ND/CEF group received ND for 19 days plus coadministration of CEF in the last 5 days. On the 19th day, the aggressive phenotypes were evaluated through the resident-intruder test. After 24 h, cerebrospinal fluid was collected to measure glutamate levels, and the pre-frontal cortex was used to assess GLT-1, pGluN2BTyr1472, and pGluN2ATyr1246 by Western blot. Synaptosomes from the left brain hemisphere was used to evaluate mitochondrial function including complex II-succinate dehydrogenase (SDH), Ca2+ handling, membrane potential (ΔѰm), and H2O2 production. ND decreased the latency for the first attack and increased the number of attacks by the resident mice against the intruder, mechanistically associated with an increase in glutamate levels and pGluN2BTyr1472 but not pGluN2ATyr1244, and GLT-1 downregulation. The abnormalities in mitochondrial Ca2+ influx, SDH, ΔѰm, and H2O2 implies in deficient energy support to the synaptic machinery. The ND/CEF group displayed a decreased aggressive behavior, normalization of glutamate and pGluN2BTyr1472levels, and mitochondrial function at synaptic terminals. In conclusion, the pharmacological modulation of GLT-1 highlights its relevance as an astrocytic target against highly impulsive and aggressive phenotypes.
Collapse
Affiliation(s)
- Marcelo S Rodolphi
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil
| | - Afonso Kopczynski
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil
| | - Randhall B Carteri
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil; Centro Universitário Metodista - Instituto Porto Alegre (IPA), Coronel Joaquim Pedro Salgado 80, Porto Alegre, RS 90420-060, Brazil
| | - Monia Sartor
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil
| | - Fernanda U Fontella
- Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil
| | - Marceli Feldmann
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil
| | - Gisele Hansel
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil; Robert A. Groff Professor of Teaching and Research in Neurosurgery Department: Neurosurgery, University of Pennsylvania, 105 Hayden Hall 3320 Smith Walk, Philadelphia, PA 19104-6316, USA; Laboratório de Neuroinflamação e Neuroimunologia, Instituto do Cérebro do Rio Grande do Sul, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Nathan R Strogulski
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil
| | - Luis V Portela
- Laboratório de Neurotrauma e Biomarcadores, Departamento de Bioquímica, Programa de Pós-Graduação Bioquímica, Universidade Federal do Rio Grande do Sul-UFRGS, Ramiro Barcelos 2600, anexo, Porto Alegre, RS 90035-003, Brazil.
| |
Collapse
|
16
|
Xian XH, Gao JX, Qi J, Fan SJ, Zhang M, Li WB. Activation of p38 MAPK participates in the sulbactam-induced cerebral ischemic tolerance mediated by glial glutamate transporter-1 upregulation in rats. Sci Rep 2020; 10:20601. [PMID: 33244020 PMCID: PMC7692545 DOI: 10.1038/s41598-020-77583-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Our previous studies have shown that sulbactam can play a neuroprotection role in hippocampal neurons by upregulating the expression and function of glial glutamate transporter-1 (GLT-1) during ischemic insult. Here, using rat global cerebral ischemia model, we studied in vivo the role of p38 mitogen-activated protein kinases (MAPK) in the sulbactam-induced GLT-1 upregulation and neuroprotection against ischemia. The hippocampal CA1 field was selected as observing target. The expressions of phosphorylated-p38 MAPK and GLT-1 were assayed with western blot analysis and immunohistochemistry. The condition of delayed neuronal death (DND) was assayed with neuropathological evaluation under thionin staining. It was shown that administration of sulbactam protected CA1 hippocampal neurons against ischemic insult accompanied with significantly upregulation in the expressions of phosphorylated-p38 MAPK and GLT-1. The time course analysis showed that sulbactam activated p38 MAPK before the GLT-1 upregulation in either normal or global cerebral ischemic rats. Furthermore, inhibiting p38 MAPK activation by SB203580 blocked the GLT-1 upregulation and neuroprotection induced by sulbactam. The above results suggested that p38 MAPK, at least partly, participated in the sulbactam-induced brain tolerance to ischemia mediated by GLT-1 upregulation in rats.
Collapse
Affiliation(s)
- Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Jun-Xia Gao
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Jie Qi
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Shu-Juan Fan
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China. .,Neuroscience Research Center of Hebei Medical University, Shijiazhuang, People's Republic of China.
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China. .,Neuroscience Research Center of Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
17
|
Gao J, Liu L, Liu C, Fan S, Liu L, Liu S, Xian XH, Li WB. GLT-1 Knockdown Inhibits Ceftriaxone-Mediated Improvements on Cognitive Deficits, and GLT-1 and xCT Expression and Activity in APP/PS1 AD Mice. Front Aging Neurosci 2020; 12:580772. [PMID: 33132901 PMCID: PMC7574737 DOI: 10.3389/fnagi.2020.580772] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/08/2020] [Indexed: 11/16/2022] Open
Abstract
Objective Glutamate transporter-1 (GLT-1) and system xc– mediate glutamate uptake and release, respectively. Ceftriaxone has been reported to upregulate GLT-1 expression and improve cognitive decline in APP/PS1 mice. The aim of the present study was to elucidate the role of GLT-1 in ceftriaxone-mediated improvement on cognitive deficits and associated changes in xCT (catalytic subunit of system xc–) expression and activity using GLT-1 knockdown APP/PS1 mice. Methods GLT-1 knockdown (GLT-1±) mice were generated in C57BL/6J mice using the CRISPR/Cas9 technique and crossed to APP/PS1 mice to generate GLT-1±APP/PS1 mice. The cognition was evaluated by novel object recognition and Morris water maze tests. GLT-1 and xCT expression, GLT-1 uptake for glutamate, and glutathione levels of hippocampus were assayed using Western blot and immunohistochemistry, 3H-glutamate, and glutathione assay kit, respectively. Results In comparison with wild-type mice, APP/PS1 mice exhibited significant cognitive deficits, represented with poor performance in novel object recognition and Morris water maze tests, downregulated GLT-1 expression and glutamate uptake. Ceftriaxone treatment significantly improved the above impairments in APP/PS1 mice, but had negligible impact in GLT-1±APP/PS1 mice. The xCT expression increased in APP/PS1 and GLT-1±APP/PS1 mice. This upregulation might be a compensatory change against the accumulated glutamate resulting from GLT-1 impairment. Ceftriaxone treatment restored xCT expression in APP/PS1 mice, but not in GLT-1±APP/PS1 mice. Glutathione levels decreased in APP/PS1 mice in comparison to the wild-type group. After ceftriaxone administration, the decline in glutathione level was restored in APP/PS1 mice, but not in GLT-1±APP/PS1 mice. Conclusion Ceftriaxone improves cognitive impairment of APP/PS1 mice by upregulating GLT-1-mediated uptake of glutamate and co-regulation of GLT-1 and xCT in APP/PS1 mice.
Collapse
Affiliation(s)
- JunXia Gao
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - LiZhe Liu
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Chao Liu
- Hebei Key Lab of Laboratory Animal Science, Laboratory Animal Center, Hebei Medical University, Shijiazhuang, China
| | - ShuJuan Fan
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - LiRong Liu
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - ShuFeng Liu
- Hebei Key Lab of Laboratory Animal Science, Laboratory Animal Center, Hebei Medical University, Shijiazhuang, China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Wen-Bin Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
18
|
Neurobiology, Functions, and Relevance of Excitatory Amino Acid Transporters (EAATs) to Treatment of Refractory Epilepsy. CNS Drugs 2020; 34:1089-1103. [PMID: 32926322 DOI: 10.1007/s40263-020-00764-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epilepsy is one of the most prevalent and devastating neurological disorders characterized by episodes of unusual sensations, loss of awareness, and reoccurring seizures. The frequency and intensity of epileptic fits can vary to a great degree, with almost a third of all cases resistant to available therapies. At present, there is a major unmet need for effective and specific therapeutic intervention. Impairments of the exquisite balance between excitatory and inhibitory synaptic processes in the brain are considered key in the onset and pathophysiology of the disease. As the primary excitatory neurotransmitter in the central nervous system, glutamate has been implicated in the process, with the glutamatergic system holding center stage in the pathobiology as well as in developing disease-modifying therapies. Emerging data pinpoint impairments of glutamate clearance as one of the key causative factors in drug-resistant disease forms. Reinstatement of glutamate homeostasis using pharmacological and genetic modulation of glutamate clearance is therefore considered to be of major translational relevance. In this article, we review the neurobiological and clinical evidence suggesting complex aberrations in the activity and functions of excitatory amino acid transporters (EAATs) in epilepsy, with knock-on effects on glutamate homeostasis as a leading cause for the development of refractory forms. We consider the emerging data on pharmacological and genetic manipulations of EAATs, with reference to seizures and glutamate dyshomeostasis, and review their fundamental and translational relevance. We discuss the most recent advances in the EAATs research in human and animal models, along with numerous questions that remain open for debate and critical appraisal. Contrary to the widely held view on EAATs as a promising therapeutic target for management of refractory epilepsy as well as other neurological and psychiatric conditions related to glutamatergic hyperactivity and glutamate-induced cytotoxicity, we stress that the true relevance of EAAT2 as a target for medical intervention remains to be fully appreciated and verified. Despite decades of research, the emerging properties and functional characteristics of glutamate transporters and their relationship with neurophysiological and behavioral correlates of epilepsy challenge the current perception of this disease and fit unambiguously in neither EAATs functional deficit nor in reversal models. We stress the pressing need for new approaches and models for research and restoration of the physiological activity of glutamate transporters and synaptic transmission to achieve much needed therapeutic effects. The complex mechanism of EAATs regulation by multiple factors, including changes in the electrochemical environment and ionic gradients related to epileptic hyperactivity, impose major therapeutic challenges. As a final note, we consider the evolving views and present a cautious perspective on the key areas of future progress in the field towards better management and treatment of refractory disease forms.
Collapse
|
19
|
Shakkour Z, Habashy KJ, Berro M, Takkoush S, Abdelhady S, Koleilat N, Eid AH, Zibara K, Obeid M, Shear D, Mondello S, Wang KK, Kobeissy F. Drug Repurposing in Neurological Disorders: Implications for Neurotherapy in Traumatic Brain Injury. Neuroscientist 2020; 27:620-649. [PMID: 33089741 DOI: 10.1177/1073858420961078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) remains a significant leading cause of death and disability among adults and children globally. To date, there are no Food and Drug Administration-approved drugs that can substantially attenuate the sequelae of TBI. The innumerable challenges faced by the conventional de novo discovery of new pharmacological agents led to the emergence of alternative paradigm, which is drug repurposing. Repurposing of existing drugs with well-characterized mechanisms of action and human safety profiles is believed to be a promising strategy for novel drug use. Compared to the conventional discovery pathways, drug repurposing is less costly, relatively rapid, and poses minimal risk of the adverse outcomes to study on participants. In recent years, drug repurposing has covered a wide range of neurodegenerative diseases and neurological disorders including brain injury. This review highlights the advances in drug repurposing and presents some of the promising candidate drugs for potential TBI treatment along with their possible mechanisms of neuroprotection. Edaravone, glyburide, ceftriaxone, levetiracetam, and progesterone have been selected due to their potential role as putative TBI neurotherapeutic agents. These drugs are Food and Drug Administration-approved for purposes other than brain injuries; however, preclinical and clinical studies have shown their efficacy in ameliorating the various detrimental outcomes of TBI.
Collapse
Affiliation(s)
- Zaynab Shakkour
- Department of Biochemistry & Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Moussa Berro
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samira Takkoush
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nadia Koleilat
- Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Makram Obeid
- Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Deborah Shear
- Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Sicilia, Italy
| | - Kevin K Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
20
|
Zhao Y, Yang J, Li C, Zhou G, Wan H, Ding Z, Wan H, Zhou H. Role of the neurovascular unit in the process of cerebral ischemic injury. Pharmacol Res 2020; 160:105103. [PMID: 32739425 DOI: 10.1016/j.phrs.2020.105103] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
Cerebral ischemic injury exhibits both high morbidity and mortality worldwide. Traditional research of the pathogenesis of cerebral ischemic injury has focused on separate analyses of the involved cell types. In recent years, the neurovascular unit (NVU) mechanism of cerebral ischemic injury has been proposed in modern medicine. Hence, more effective strategies for the treatment of cerebral ischemic injury may be provided through comprehensive analysis of brain cells and the extracellular matrix. However, recent studies that have investigated the function of the NVU in cerebral ischemic injury have been insufficient. In addition, the metabolism and energy conversion of the NVU depend on interactions among multiple cell types, which make it difficult to identify the unique contribution of each cell type. Therefore, in the present review, we comprehensively summarize the regulatory effects and recovery mechanisms of four major cell types (i.e., astrocytes, microglia, brain-microvascular endothelial cells, and neurons) in the NVU under cerebral ischemic injury, as well as discuss the interactions among these cell types in the NVU. Furthermore, we discuss the common signaling pathways and signaling factors that mediate cerebral ischemic injury in the NVU, which may help to provide a theoretical basis for the comprehensive elucidation of cerebral ischemic injury.
Collapse
Affiliation(s)
- Yu Zhao
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Guoying Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haofang Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Zhishan Ding
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
21
|
Smaga I, Fierro D, Mesa J, Filip M, Knackstedt LA. Molecular changes evoked by the beta-lactam antibiotic ceftriaxone across rodent models of substance use disorder and neurological disease. Neurosci Biobehav Rev 2020; 115:116-130. [PMID: 32485268 DOI: 10.1016/j.neubiorev.2020.05.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
Ceftriaxone is a beta-lactam antibiotic that increases the expression of the major glutamate transporter, GLT-1. As such, ceftriaxone ameliorates symptoms across multiple rodent models of neurological diseases and substance use disorders. However, the mechanism behind GLT-1 upregulation is unknown. The present review synthesizes this literature in order to identify commonalities in molecular changes. We find that ceftriaxone (200 mg/kg for at least two days) consistently restores GLT-1 expression in multiple rodent models of neurological disease, especially when GLT-1 is decreased in the disease model. The same dose given to healthy/drug-naive rodents does not reliably upregulate GLT-1 in any brain region except the hippocampus. Increased GLT-1 expression does not consistently arise from transcriptional regulation, and is likely to be due to trafficking changes. In addition to altered transporter expression, ceftriaxone ameliorates neuropathologies (e.g. tau, amyloid beta, cell death) and aberrant protein expression associated with a number of neurological disease models. Taken together, these results indicate that ceftriaxone remains a strong candidate for treatment of multiple disorders in the clinic.
Collapse
Affiliation(s)
- Irena Smaga
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL, 31-343, Kraków, Poland
| | - Daniel Fierro
- Department of Psychology, University of Florida, 945 Center Dr., Gainesville, FL, 32611, USA
| | - Javier Mesa
- Department of Psychology, University of Florida, 945 Center Dr., Gainesville, FL, 32611, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32611, USA
| | - Malgorzata Filip
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL, 31-343, Kraków, Poland
| | - Lori A Knackstedt
- Department of Psychology, University of Florida, 945 Center Dr., Gainesville, FL, 32611, USA; Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
22
|
Amyloid Fibril-Induced Astrocytic Glutamate Transporter Disruption Contributes to Complement C1q-Mediated Microglial Pruning of Glutamatergic Synapses. Mol Neurobiol 2020; 57:2290-2300. [PMID: 32008166 DOI: 10.1007/s12035-020-01885-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/21/2020] [Indexed: 01/23/2023]
Abstract
The complement C1q plays a critical role in microglial phagocytosis of glutamatergic synapses and in the pathogenesis of neuroinflammation in Alzheimer's disease (AD). We recently reported that upregulation of metabotropic glutamate receptor signaling is associated with increased synaptic C1q production and subsequent microglial phagocytosis of synapses in the rodent models of AD. Here, we explored the role of astrocytic glutamate transporter in the synaptic C1q production and microglial phagocytosis of hippocampal glutamatergic synapses in a rat model of AD. Activation of astrocyte and reduction glutamate transporter 1 (GLT1) were noted after bilateral microinjection of amyloid-beta (Aβ1-40) fibrils into the hippocampal CA1 area of rats. Ceftriaxone is a β-lactam antibiotic that upregulates GLT1 expression. Bilateral microinjection of ceftriaxone recovered GLT1 expression, decreased synaptic C1q production, suppressed microglial phagocytosis of glutamatergic synapses in the hippocampal CA1, and attenuated synaptic and cognitive deficits in rats microinjected with Aβ1-40. In contrast, artificial suppression of GLT1 activity by DL-threo-beta-benzyloxyaspartate (DL-TBOA) in naïve rats induced synaptic C1q expression and microglial phagocytosis of glutamatergic synapses in the hippocampal CA1 area, resulting in synaptic and cognitive dysfunction. These findings demonstrated that impairment of astrocytic glutamate transporter plays a role in the pathogenesis of AD.
Collapse
|
23
|
Pais-Vieira M, Kunicki C, Peres A, Sousa N. Ceftriaxone modulates the acute corticosterone effects in local field potentials in the primary somatosensory cortex of anesthetized mice. Sci Rep 2019; 9:20289. [PMID: 31889134 PMCID: PMC6937346 DOI: 10.1038/s41598-019-56827-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Stress responses are associated with elevations in corticosterone levels and, as a consequence, increases in glutamate in the central nervous system which can lead to neurological impairment. Ceftriaxone promotes glutamate transport and has been used to reduce glutamate toxicity, but so far it is not known whether ceftriaxone is able to reverse the effects of corticosterone administration. Here we describe the separate and combined effects of acute ceftriaxone and acute corticosterone administration in local field potentials (LFPs) recorded from the somatosensory cortex (S1) of anesthetized mice. For this, LFPs were recorded from groups of anesthetized mice injected with saline, corticosterone, ceftriaxone, or both. Comparison of global state maps, and their displacements, as measured by ratios of different frequency bands (Ratio 1: 0.5–20 Hz/0.5–45 Hz; and Ratio 2: 0.5–4.5 Hz/0.5–9 Hz) revealed distinct and opposite effects for corticosterone and for ceftriaxone. Corticosterone specifically increased the displacement in Ratio 2, while ceftriaxone decreased it; in addition, when both corticosterone and ceftriaxone were injected, Ratio 2 displacement values were again similar to those of the control group. The present results suggest that ceftriaxone and corticosterone modulate specific frequency bands in opposite directions and reveal a potential role for ceftriaxone in counteracting the effects of corticosterone.
Collapse
Affiliation(s)
- Miguel Pais-Vieira
- Center for Interdisciplinary Research in Health, Institute of Health Sciences, Universidade Católica Portuguesa, Porto, Portugal. .,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal. .,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, 4710-057, Portugal. .,Clinical Academic Center (2CA-Braga), Braga, Portugal.
| | - Carolina Kunicki
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaiba, Brazil
| | - André Peres
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaiba, Brazil
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, 4710-057, Portugal.,Clinical Academic Center (2CA-Braga), Braga, Portugal
| |
Collapse
|
24
|
Fan S, Xian X, Li L, Yao X, Hu Y, Zhang M, Li W. Ceftriaxone Improves Cognitive Function and Upregulates GLT-1-Related Glutamate-Glutamine Cycle in APP/PS1 Mice. J Alzheimers Dis 2019; 66:1731-1743. [PMID: 30452416 DOI: 10.3233/jad-180708] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is characterized by progressive impairment of learning, memory, and cognitive deficits. Glutamate is the major excitatory neurotransmitter in the central nervous system and plays an important role in learning, memory, and cognition. The homeostasis and reutilization of glutamate are dependent on astrocytic uptake by glutamate transporter-1 (GLT-1) and the subsequent glutamate-glutamine cycle. Increasing evidence showed impairments in GLT-1 expression and uptake activity and glutamate-glutamine cycle in AD. Ceftriaxone (Cef) has been reported to upregulate the expression and uptake of GLT-1. Therefore, the present study was undertaken to explore whether Cef can improve cognitive deficits of APP/PS1 mice in early stage of AD by upregulating GLT-1 expression, and then promoting the glutamate-glutamine cycle. It was shown that Cef treatment significantly alleviated the cognitive deficits measured by Morris water maze test and upregulated GLT-1 protein expression in the hippocampus of APP/PS1 mice. Particularly, the activity of glutamine synthetase (GS) and the protein expression of system N glutamine transporter 1 (SN1), which are the key factors involved in the glutamate-glutamine cycle, were significantly upregulated as well after the Cef treatment. Furthermore, inhibition of GLT-1 uptake activity by dihydrokainic acid, an inhibitor of GLT-1, blocked the Cef-induced improvement on the cognitive deficits, GS activity, and SN1 expression. The above results suggested that Cef could improve cognitive deficits of APP/PS1 mice in early stage of AD by upregulating the GLT-1 expression, GS activity, and SN1 expression, which would lead to stimulating the glutamate-glutamine cycle.
Collapse
Affiliation(s)
- ShuJuan Fan
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - XiaoHui Xian
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - Li Li
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - XiaoGuang Yao
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - YuYan Hu
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - Min Zhang
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China
| | - WenBin Li
- Department of Pathophysiology, Neuroscience Center, Hebei Medical University, Shijiazhuang, P.R. China.,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, P.R. China
| |
Collapse
|
25
|
Ceftriaxone Treatment Affects EAAT2 Expression and Glutamatergic Neurotransmission and Exerts a Weak Anticonvulsant Effect in Young Rats. Int J Mol Sci 2019; 20:ijms20235852. [PMID: 31766528 PMCID: PMC6928884 DOI: 10.3390/ijms20235852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is a common neurological disorder. Despite the availability of a wide range of antiepileptic drugs, these are unsuccessful in preventing seizures in 20–30% of patients. Therefore, new pharmacological strategies are urgently required to control seizures. Modulation of glutamate uptake may have potential in the treatment of pharmacoresistant forms of epilepsy. Previous research showed that the antibiotic ceftriaxone (CTX) increased the expression and functional activity of excitatory amino acid transporter 2 (EAAT2) and exerted considerable anticonvulsant effects. However, other studies did not confirm a significant anticonvulsant effect of CTX administration. We investigated the impacts of CTX treatment on EAAT expression and glutamatergic neurotransmission, as well its anticonvulsant action, in young male Wistar rats. As shown by a quantitative real-time polymerase chain reaction (qPCR) assay and a Western blot analysis, the mRNA but not the protein level of EAAT2 increased in the hippocampus following CTX treatment. Repetitive CTX administration had only a mild anticonvulsant effect on pentylenetetrazol (PTZ)-induced convulsions in a maximal electroshock threshold test (MEST). CTX treatment did not affect the glutamatergic neurotransmission, including synaptic efficacy, short-term facilitation, or the summation of excitatory postsynaptic potentials (EPSPs) in the hippocampus and temporal cortex. However, it decreased the field EPSP (fEPSP) amplitudes evoked by intense electrical stimulation. In conclusion, in young rats, CTX treatment did not induce overexpression of EAAT2, therefore exerting only a weak antiseizure effect. Our data provide new insight into the effects of modulation of EAAT2 expression on brain functioning.
Collapse
|
26
|
Blaker AL, Moore ER, Yamamoto BK. Serial exposure to ethanol drinking and methamphetamine enhances glutamate excitotoxicity. J Neurochem 2019; 151:749-763. [PMID: 31478210 DOI: 10.1111/jnc.14861] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/05/2019] [Accepted: 08/26/2019] [Indexed: 12/22/2022]
Abstract
A significant comorbidity exists between alcohol and methamphetamine (Meth) abuse but the neurochemical consequences of this co-abuse are unknown. Alcohol and Meth independently and differentially affect glutamatergic transmission but the unique effects of their serial exposure on glutamate signaling in mediating damage to dopamine neurons are unknown. Sprague-Dawley rats had intermittent voluntary access to 10% ethanol (EtOH) every other day and water over 28 days and were then administered a binge injection regimen of Meth or saline. EtOH drinking decreased the glutamate aspartate transporter and increased basal extracellular concentrations of glutamate within the striatum when measured after the last day of drinking. Ceftriaxone is known to increase the expression and/or activity of glutamate transporters in the brain and prevented both the decreases in glutamate aspartate transporter and the increases in basal extracellular glutamate when administered during EtOH drinking. EtOH drinking also exacerbated the acute increases in extracellular glutamate observed upon Meth exposure, the subsequent increases in spectrin proteolysis, and the long-term decreases in dopamine content in the striatum, all of which were attenuated by ceftriaxone administration during EtOH drinking only. These results implicate EtOH-induced increases in extracellular glutamate and corresponding decreases in glutamate uptake as mechanisms that contribute to the vulnerability produced by EtOH drinking and the unique neurotoxicity observed after serial exposure to Meth that is not observed with either drug alone. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Amanda L Blaker
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Elizabeth R Moore
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bryan K Yamamoto
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
27
|
Yimer EM, Hishe HZ, Tuem KB. Repurposing of the β-Lactam Antibiotic, Ceftriaxone for Neurological Disorders: A Review. Front Neurosci 2019; 13:236. [PMID: 30971875 PMCID: PMC6444273 DOI: 10.3389/fnins.2019.00236] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
To date, there is no cure or disease-modifying agents available for most well-known neurological disorders. Current therapy is typically focused on relieving symptoms and supportive care in improving the quality of life of affected patients. Furthermore, the traditional de novo drug discovery technique is more challenging, particularly for neurological disorders. Therefore, the repurposing of existing drugs for these conditions is believed to be an efficient and dynamic approach that can substantially reduce the investments spent on drug development. Currently, there is emerging evidence that suggests the potential effect of a beta-lactam antibiotic, ceftriaxone (CEF), to alleviate the symptoms of different experimentally-induced neurological disorders: Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, epileptic-seizure, brain ischemia, traumatic brain injuries, and neuropathic pain. CEF also affects the markers of oxidative status and neuroinflammation, glutamatergic systems as well as various aggregated toxic proteins involved in the pathogenesis of different neurological disorders. Moreover, it was found that CEF administration to drug dependent animal models improved the withdrawal symptoms upon drug discontinuation. Thus, this review aimed to describe the effects of CEF against multiple models of neurological illnesses, drug dependency, and withdrawal. It also emphasizes the possible mechanisms of neuroprotective actions of CEF with respective neurological maladies.
Collapse
Affiliation(s)
- Ebrahim M Yimer
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Hailemichael Zeru Hishe
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Kald Beshir Tuem
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
28
|
Chronic Unexpected Mild Stress Destroys Synaptic Plasticity of Neurons through a Glutamate Transporter, GLT-1, of Astrocytes in the Ischemic Stroke Rat. Neural Plast 2019; 2019:1615925. [PMID: 31019528 PMCID: PMC6452568 DOI: 10.1155/2019/1615925] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/27/2019] [Accepted: 02/21/2019] [Indexed: 02/03/2023] Open
Abstract
Background and Objective Chronic unexpected mild stress (CUMS) destroys synaptic plasticity of hippocampal regenerated neurons that may be involved in the occurrence of poststroke depression. Astrocytes uptake glutamate at the synapse and provide metabolic support for neighboring neurons. Currently, we aim to investigate whether CUMS inhibits synaptic formation of regenerated neurons through a glutamate transporter, GLT-1, of astrocytes in the ischemic stroke rats. Method We exposed the ischemic stroke rats to ceftriaxone, during the CUMS intervention period to determine the effects of GLT-1 on glutamate circulation by immunofluorescence and mass spectrometry and its influences to synaptic plasticity by western blot and transmission electron microscopy. Result CUMS evidently reduced the level of astroglial GLT-1 in the hippocampus of the ischemic rats (p < 0.05), resulting in smaller amount of glutamate being transported into astrocytes surrounding synapses (p < 0.05), and then expression of synaptophysin was suppressed (p < 0.05) in hippocampal dentate gyrus. The ultrastructures of synapses in dentate gyrus were adversely influenced including decreased proportion of smile synapses, shortened thickness of postsynaptic density, reduced number of vesicles, and widened average distance of the synaptic cleft (all p < 0.05). Moreover, ceftriaxone can promote glutamate circulation and synaptic plasticity (all p < 0.05) by raising astroglial GLT-1 (p < 0.05) and then improve depressive behaviors of the CUMS-induced model rats (p < 0.05). Conclusion Our study shows that CUMS destroys synaptic plasticity of regenerated neurons in the hippocampus through a glutamate transporter, GLT-1, of astrocytes in the ischemic stroke rats. This may indicate one potential pathogenesis of poststroke depression.
Collapse
|
29
|
The mechanism of GLT-1 mediating cerebral ischemic injury depends on the activation of p38 MAPK. Brain Res Bull 2019; 147:1-13. [PMID: 30731111 DOI: 10.1016/j.brainresbull.2019.01.028] [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: 11/03/2018] [Revised: 01/01/2019] [Accepted: 01/29/2019] [Indexed: 12/15/2022]
Abstract
The previous studies have shown that glial glutamate transporter-1 (GLT-1) participates in cerebral ischemic injury in rats. However, the mechanism involved remains to be elucidated. This study was undertaken to investigate whether p38 MAPK was involved in regulating GLT-1 in the process. At first, it was observed that global brain ischemia for 8 min led to obvious delayed neuronal death, GLT-1 down-regulation and p-p38 MAPK up-regulation in CA1 hippocampus in rats. Then, whether p-p38 MAPK was involved in regulating GLT-1 during cerebral ischemic injury was studied in vitro. Astrocyte-neuron co-cultures exposed to oxygen and glucose deprivation (OGD) were used to mimic brain ischemia. It was observed that lethal OGD (4-h OGD) decreased GLT-1 expression and increased p-p38 MAPK expression in astrocytes. The p-p38 MAPK protein rised from 0 min to 48 h that is the end time of the observation, and the peak value was at 12 h, which was 12.45 times of the control group. Moreover, pre-administration of p38 MAPK inhibitor SB203580 or its siRNA dose-dependently increased GLT-1 expression, and meanwhile alleviated the neuronal death induced by lethal OGD. The above results indicated that p38 MAPK signaling pathway participated in regulating GLT-1 during OGD injury in vitro. Finally, back to in vivo experiment, it was found that pre-administration of SB203580 by intracerebroventricular injection dose-dependently reversed the down-regulation of GLT-1 expression and attenuated the delayed neuronal death normally induced by global brain ischemia in CA1 hippocampus in rats. Taken together, it can be concluded that the mechanism of GLT-1 mediating cerebral ischemic injury depends on the activation of p38 MAPK.
Collapse
|
30
|
Zhang LN, Hao L, Guo YS, Wang HY, Li LL, Liu LZ, Li WB. Are glutamate transporters neuroprotective or neurodegenerative during cerebral ischemia? J Mol Med (Berl) 2019; 97:281-289. [PMID: 30675649 DOI: 10.1007/s00109-019-01745-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/10/2019] [Accepted: 01/14/2019] [Indexed: 12/14/2022]
Abstract
The accumulation of glutamate (Glu) in the synaptic cleft during cerebral ischemia triggers the death of neurons, causing mental or physical handicap. However, the mechanisms of the alteration in Glu homeostasis and the imbalance between the release and clearance of Glu in ischemia are not yet completely understood. Additionally, the role of Glu transporters in regulating Glu concentration in the synaptic cleft is controversial. This review aims to provide readers with an in-depth understanding of Glu transporters in the early or later stages of ischemic events, or in mild or severe cerebral ischemia via alteration of Glu transporter expression, reversal of Glu transporters function, and trafficking between membrane and cytoplasm, to further clarify whether the Glu transporters are neuroprotective or neurodegenerative during cerebral ischemia. We provide the insights for deeper understanding of the mechanism of Glu transporters regulation after different periods and severities of cerebral ischemia.
Collapse
Affiliation(s)
- Li-Nan Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, China
| | - Liang Hao
- Department of Neurosurgery, Third Hospital of Shijiazhuang, Beijing, 050011, Hebei, China
| | - Yu-Song Guo
- Department of Traumatology, Third Hospital of Shijiazhuang, Beijing, 050011, Hebei, China
| | - Hai-Yan Wang
- Pharmaceutical Preparation Section, Third Hospital of Shijiazhuang, Beijing, 050011, Hebei, China
| | - Lin-Lin Li
- Clinical Medicine, College of Basic Medicine, Hebei Medical University, Beijing, 050017, Hebei, China
| | - Li-Zhe Liu
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
31
|
Yu D, Cheng Z, Ali AI, Wang J, Le K, Chibaatar E, Guo Y. Down-expressed GLT-1 in PSD astrocytes inhibits synaptic formation of NSC-derived neurons in vitro. Cell Cycle 2018; 18:105-114. [PMID: 30558468 DOI: 10.1080/15384101.2018.1560201] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Little is known about the effect of astroglial GLT-1 of post-stroke depression (PSD) rat model on the function of neural stem cells (NSCs). This study aimed to investigate whether astroglial GLT-1 of PSD rats affect differentiation of NSCs from neonatal rat hippocampus and synaptic formation of NSC-derived neurons. Astrocytes were isolated from the left hippocampus of normal adult SD rats and PSD rats. A lentiviral vector was used to silence the expression of GLT-1 in astrocytes of PSD rats. NSCs were respectively co-cultured with normal (control), PSD, and GLT-1 silenced astrocytes for 7 days. GLT-1, GFAP, MAP2, Synaptophysin (SYN), glutamate (Glu) and glutamine (Gln) were respectively measured by qRT-PCR, western blot, immunofluorescence and efficient mass spectrometry (MS). PSD astrocytes increased the number of NSC-derived astrocytes, but inhibited the expression of GLT-1 of NSC-derived astrocytes and synapses of NSC-derived neurons. On the basis of the low expression of GLT-1 in PSD astrocytes, we further silenced GLT-1 in PSD astrocytes. Interestingly, GLT-1 silenced PSD astrocytes more obviously inhibited synapses of NSC-derived neurons, but increased the number of NSC-derived neurons and reversed the expression of GLT-1 in NSC-derived astrocytes. At the same time, concentration of glutamate in the medium elevated, and glutamine in the medium gradually reduced. In NSC-derived neurons and astrocytes, glutamate metabolism was also affected by changed GLT-1. Down-expressed GLT-1 in PSD astrocytes stimulated NSCs differentiating into astrocytes, but inhibiting the formation of functional synapses by influencing glutamate metabolism in vitro.
Collapse
Affiliation(s)
- Dafan Yu
- a Department of Neurology Affiliated , Zhongda Hospital of Southeast University , Nanjing , Jiangsu Province , China.,b School of Medicine , Southeast University , Nanjing , Jiangsu Province , China
| | - Zhenxing Cheng
- b School of Medicine , Southeast University , Nanjing , Jiangsu Province , China
| | - Abdoulaye Idriss Ali
- a Department of Neurology Affiliated , Zhongda Hospital of Southeast University , Nanjing , Jiangsu Province , China.,b School of Medicine , Southeast University , Nanjing , Jiangsu Province , China
| | - Jiamin Wang
- a Department of Neurology Affiliated , Zhongda Hospital of Southeast University , Nanjing , Jiangsu Province , China.,b School of Medicine , Southeast University , Nanjing , Jiangsu Province , China
| | - Kai Le
- a Department of Neurology Affiliated , Zhongda Hospital of Southeast University , Nanjing , Jiangsu Province , China.,b School of Medicine , Southeast University , Nanjing , Jiangsu Province , China
| | - Enkhmurun Chibaatar
- a Department of Neurology Affiliated , Zhongda Hospital of Southeast University , Nanjing , Jiangsu Province , China.,b School of Medicine , Southeast University , Nanjing , Jiangsu Province , China
| | - Yijing Guo
- a Department of Neurology Affiliated , Zhongda Hospital of Southeast University , Nanjing , Jiangsu Province , China.,b School of Medicine , Southeast University , Nanjing , Jiangsu Province , China
| |
Collapse
|
32
|
Ketamine and Ceftriaxone-Induced Alterations in Glutamate Levels Do Not Impact the Specific Binding of Metabotropic Glutamate Receptor Subtype 5 Radioligand [ 18F]PSS232 in the Rat Brain. Pharmaceuticals (Basel) 2018; 11:ph11030083. [PMID: 30158438 PMCID: PMC6161118 DOI: 10.3390/ph11030083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/23/2018] [Accepted: 08/25/2018] [Indexed: 01/21/2023] Open
Abstract
Several studies showed that [11C]ABP688 binding is altered following drug-induced perturbation of glutamate levels in brains of humans, non-human primates and rats. We evaluated whether the fluorinated derivative [18F]PSS232 can be used to assess metabotropic glutamate receptor 5 (mGluR5) availability in rats after pharmacological challenge with ketamine, known to increase glutamate, or ceftriaxone, known to decrease glutamate. In vitro autoradiography was performed on rat brain slices with [18F]PSS232 to prove direct competition of the drugs for mGluR5. One group of rats were challenged with a bolus injection of either vehicle, racemic ketamine, S-ketamine or ceftriaxone followed by positron emission tomography PET imaging with [18F]PSS232. The other group received an infusion of the drugs during the PET scan. Distribution volume ratios (DVRs) were calculated using a reference tissue model. In vitro autoradiography showed no direct competition of the drugs with [18F]PSS232 for the allosteric binding site of mGluR5. DVRs of [18F]PSS232 binding in vivo did not change in any brain region neither after bolus injection nor after infusion. We conclude that [18F]PSS232 has utility for measuring mGluR5 density or occupancy of the allosteric site in vivo, but it cannot be used to measure in vivo fluctuations of glutamate levels in the rat brain.
Collapse
|
33
|
Qi J, Xian XH, Li L, Zhang M, Hu YY, Zhang JG, Li WB. Sulbactam Protects Hippocampal Neurons Against Oxygen-Glucose Deprivation by Up-Regulating Astrocytic GLT-1 via p38 MAPK Signal Pathway. Front Mol Neurosci 2018; 11:281. [PMID: 30158854 PMCID: PMC6104165 DOI: 10.3389/fnmol.2018.00281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022] Open
Abstract
Sulbactam is an atypical β-lactam medication and reported to be neuroprotective by up-regulating glial glutamate transporter-1 (GLT-1) in rats. The present study was undertaken to study the role of p38 MAPK signal pathway in sulbactam induced up-regulation of GLT-1 expression in astrocytes and anti-ischemic effect. Neuron-astrocyte co-cultures and astrocyte cultures from neonatal Wistar rats were used. Cerebral ischemia was mimicked by oxygen-glucose deprivation (OGD). Hoechst (HO)/propidium iodide (PI) double fluorescence staining and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay were used to evaluate neuronal death and cell viability, respectively. Immunocytochemistry and Western blot were used to detect protein expressions. Sulbactam pre-incubation significantly and dose-dependently prevented neuronal death and decline in cell viability induced by OGD in neuron-astrocyte co-cultures, and upregulated GLT-1 expression in astrocyte cultures endured OGD, which suggested that sulbactam might protect neurons against OGD by up-regulating astrocytic GLT-1 expression. It was further shown that the phosphorylated-p38 MAPK expression in astrocytes was up-regulated after the sulbactam pre-incubation and this up-regulation was moderate in amplitude. Especially, the time course of the up-regulation of phosphorylated-p38 MAPK was obviously earlier than that of GLT-1, which suggested possibility that p38 MAPK might be an upstream signal for GLT-1 up-regulation induced by sulbactam. We further found that SB203580, the specific inhibitor of p38 MAPK, dose-dependently inhibited the GLT-1 up-regulation induced by sulbactam either in non- or OGD-treated astrocytes and the protective effect of sulbactam on co-cultured neurons against OGD. Taken together, it might be concluded that sulbactam protects cerebral neurons against OGD by up-regulating astrocytic GLT-1 expression via p38 MAPK signal pathway.
Collapse
Affiliation(s)
- Jie Qi
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Li Li
- Department of Science and Technology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China.,Neuroscience Center, Hebei Medical University, Shijiazhuang, China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China.,Neuroscience Center, Hebei Medical University, Shijiazhuang, China.,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, China
| |
Collapse
|
34
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|
35
|
Krzyżanowska W, Pomierny B, Bystrowska B, Pomierny-Chamioło L, Filip M, Budziszewska B, Pera J. Ceftriaxone- and N-acetylcysteine-induced brain tolerance to ischemia: Influence on glutamate levels in focal cerebral ischemia. PLoS One 2017; 12:e0186243. [PMID: 29045497 PMCID: PMC5646803 DOI: 10.1371/journal.pone.0186243] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/27/2017] [Indexed: 01/22/2023] Open
Abstract
One of the major players in the pathophysiology of cerebral ischemia is disrupted homeostasis of glutamatergic neurotransmission, resulting in elevated extracellular glutamate (Glu) concentrations and excitotoxicity-related cell death. In the brain, Glu concentrations are regulated by Glu transporters, including Glu transporter-1 (GLT-1) and cystine/Glu antiporter (system xc-). Modulation of these transporters by administration of ceftriaxone (CEF, 200 mg/kg, i.p.) or N-acetylcysteine (NAC, 150 mg/kg, i.p.) for 5 days before focal cerebral ischemia may induce brain tolerance to ischemia by significantly limiting stroke-related damage and normalizing Glu concentrations. In the present study, focal cerebral ischemia was induced by 90-minute middle cerebral artery occlusion (MCAO). We compared the effects of CEF and NAC pretreatment on Glu concentrations in extracellular fluid and cellular-specific expression of GLT-1 and xCT with the effects of two reference preconditioning methods, namely, ischemic preconditioning and chemical preconditioning in rats. Both CEF and NAC significantly reduced Glu levels in the frontal cortex and hippocampus during focal cerebral ischemia, and this decrease was comparable with the Glu level achieved with the reference preconditioning strategies. The results of immunofluorescence staining of GLT-1 and xCT on astrocytes, neurons and microglia accounted for the observed changes in extracellular Glu levels to a certain extent. Briefly, after MCAO, the expression of GLT-1 on astrocytes decreased, but pretreatment with CEF seemed to prevent this downregulation. In addition, every intervention used in this study seemed to reduce xCT expression on astrocytes and neurons. The results of this study indicate that modulation of Glu transporter expression may restore Glu homeostasis. Moreover, our results suggest that CEF and NAC may induce brain tolerance to ischemia by influencing GLT-1 and system xc- expression levels. These transporters are presumably good targets for the development of novel therapies for brain ischemia.
Collapse
Affiliation(s)
- Weronika Krzyżanowska
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Bartosz Pomierny
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
- * E-mail:
| | - Beata Bystrowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Lucyna Pomierny-Chamioło
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Małgorzata Filip
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Drug Addiction Pharmacology, Kraków, Poland
| | - Bogusława Budziszewska
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Joanna Pera
- Department of Neurology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
36
|
Drugs to Alter Extracellular Concentration of Glutamate: Modulators of Glutamate Uptake Systems. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-1-4939-7228-9_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
37
|
Lee ML, Martinez-Lozada Z, Krizman EN, Robinson MB. Brain endothelial cells induce astrocytic expression of the glutamate transporter GLT-1 by a Notch-dependent mechanism. J Neurochem 2017; 143:489-506. [PMID: 28771710 DOI: 10.1111/jnc.14135] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 01/13/2023]
Abstract
Neuron-secreted factors induce astrocytic expression of the glutamate transporter, GLT-1 (excitatory amino acid transporter 2). In addition to their elaborate anatomic relationships with neurons, astrocytes also have processes that extend to and envelop the vasculature. Although previous studies have demonstrated that brain endothelia contribute to astrocyte differentiation and maturation, the effects of brain endothelia on astrocytic expression of GLT-1 have not been examined. In this study, we tested the hypothesis that endothelia induce expression of GLT-1 by co-culturing astrocytes from mice that utilize non-coding elements of the GLT-1 gene to control expression of reporter proteins with the mouse endothelial cell line, bEND.3. We found that endothelia increased steady state levels of reporter and GLT-1 mRNA/protein. Co-culturing with primary rat brain endothelia also increases reporter protein, GLT-1 protein, and GLT-1-mediated glutamate uptake. The Janus kinase/signal transducer and activator of transcription 3, bone morphogenic protein/transforming growth factor β, and nitric oxide pathways have been implicated in endothelia-to-astrocyte signaling; we provide multiple lines of evidence that none of these pathways mediate the effects of endothelia on astrocytic GLT-1 expression. Using transwells with a semi-permeable membrane, we demonstrate that the effects of the bEND.3 cell line are dependent upon contact. Notch has also been implicated in endothelia-astrocyte signaling in vitro and in vivo. The first step of Notch signaling requires cleavage of Notch intracellular domain by γ-secretase. We demonstrate that the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester blocks endothelia-induced increases in GLT-1. We show that the levels of Notch intracellular domain are higher in nuclei of astrocytes co-cultured with endothelia, an effect also blocked by N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester. Finally, infection of co-cultures with shRNA directed against recombination signal binding protein for immunoglobulin kappa J, a Notch effector, also reduces endothelia-dependent increases in enhanced green fluorescent protein and GLT-1. Together, these studies support a novel role for Notch in endothelia-dependent induction of GLT-1 expression. Cover Image for this issue: doi. 10.1111/jnc.13825.
Collapse
Affiliation(s)
- Meredith L Lee
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zila Martinez-Lozada
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth N Krizman
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael B Robinson
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
38
|
Lee ML, Martinez-Lozada Z, Krizman EN, Robinson MB. Brain endothelial cells induce astrocytic expression of the glutamate transporter GLT-1 by a Notch-dependent mechanism. J Neurochem 2017. [PMID: 28771710 DOI: 10.1111/jnc.13825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neuron-secreted factors induce astrocytic expression of the glutamate transporter, GLT-1 (excitatory amino acid transporter 2). In addition to their elaborate anatomic relationships with neurons, astrocytes also have processes that extend to and envelop the vasculature. Although previous studies have demonstrated that brain endothelia contribute to astrocyte differentiation and maturation, the effects of brain endothelia on astrocytic expression of GLT-1 have not been examined. In this study, we tested the hypothesis that endothelia induce expression of GLT-1 by co-culturing astrocytes from mice that utilize non-coding elements of the GLT-1 gene to control expression of reporter proteins with the mouse endothelial cell line, bEND.3. We found that endothelia increased steady state levels of reporter and GLT-1 mRNA/protein. Co-culturing with primary rat brain endothelia also increases reporter protein, GLT-1 protein, and GLT-1-mediated glutamate uptake. The Janus kinase/signal transducer and activator of transcription 3, bone morphogenic protein/transforming growth factor β, and nitric oxide pathways have been implicated in endothelia-to-astrocyte signaling; we provide multiple lines of evidence that none of these pathways mediate the effects of endothelia on astrocytic GLT-1 expression. Using transwells with a semi-permeable membrane, we demonstrate that the effects of the bEND.3 cell line are dependent upon contact. Notch has also been implicated in endothelia-astrocyte signaling in vitro and in vivo. The first step of Notch signaling requires cleavage of Notch intracellular domain by γ-secretase. We demonstrate that the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester blocks endothelia-induced increases in GLT-1. We show that the levels of Notch intracellular domain are higher in nuclei of astrocytes co-cultured with endothelia, an effect also blocked by N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester. Finally, infection of co-cultures with shRNA directed against recombination signal binding protein for immunoglobulin kappa J, a Notch effector, also reduces endothelia-dependent increases in enhanced green fluorescent protein and GLT-1. Together, these studies support a novel role for Notch in endothelia-dependent induction of GLT-1 expression. Cover Image for this issue: doi. 10.1111/jnc.13825.
Collapse
Affiliation(s)
- Meredith L Lee
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zila Martinez-Lozada
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth N Krizman
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael B Robinson
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Liu B, Teschemacher AG, Kasparov S. Astroglia as a cellular target for neuroprotection and treatment of neuro-psychiatric disorders. Glia 2017; 65:1205-1226. [PMID: 28300322 PMCID: PMC5669250 DOI: 10.1002/glia.23136] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/12/2022]
Abstract
Astrocytes are key homeostatic cells of the central nervous system. They cooperate with neurons at several levels, including ion and water homeostasis, chemical signal transmission, blood flow regulation, immune and oxidative stress defense, supply of metabolites and neurogenesis. Astroglia is also important for viability and maturation of stem-cell derived neurons. Neurons critically depend on intrinsic protective and supportive properties of astrocytes. Conversely, all forms of pathogenic stimuli which disturb astrocytic functions compromise neuronal functionality and viability. Support of neuroprotective functions of astrocytes is thus an important strategy for enhancing neuronal survival and improving outcomes in disease states. In this review, we first briefly examine how astrocytic dysfunction contributes to major neurological disorders, which are traditionally associated with malfunctioning of processes residing in neurons. Possible molecular entities within astrocytes that could underpin the cause, initiation and/or progression of various disorders are outlined. In the second section, we explore opportunities enhancing neuroprotective function of astroglia. We consider targeting astrocyte-specific molecular pathways which are involved in neuroprotection or could be expected to have a therapeutic value. Examples of those are oxidative stress defense mechanisms, glutamate uptake, purinergic signaling, water and ion homeostasis, connexin gap junctions, neurotrophic factors and the Nrf2-ARE pathway. We propose that enhancing the neuroprotective capacity of astrocytes is a viable strategy for improving brain resilience and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Beihui Liu
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
| | - Anja G. Teschemacher
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
| | - Sergey Kasparov
- School of Physiology, Pharmacology and NeuroscienceUniversity of Bristol, University WalkBS8 1TDUnited Kingdom
- Institute for Chemistry and BiologyBaltic Federal UniversityKaliningradRussian Federation
| |
Collapse
|
40
|
Interplay between mitochondrial metabolism and oxidative stress in ischemic stroke: An epigenetic connection. Mol Cell Neurosci 2017; 82:176-194. [DOI: 10.1016/j.mcn.2017.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/26/2017] [Accepted: 05/24/2017] [Indexed: 12/18/2022] Open
|
41
|
Chelini A, Brogi S, Paolino M, Di Capua A, Cappelli A, Giorgi G, Farzad M, Di Cesare Mannelli L, Micheli L, Ghelardini C, Anzini M. Synthesis and Biological Evaluation of Novel Neuroprotective Pyridazine Derivatives as Excitatory Amino Acid Transporter 2 (EAAT2) Activators. J Med Chem 2017; 60:5216-5221. [DOI: 10.1021/acs.jmedchem.7b00383] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Alessia Chelini
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro
2, 53100 Siena, Italy
| | - Simone Brogi
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro
2, 53100 Siena, Italy
| | - Marco Paolino
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro
2, 53100 Siena, Italy
| | - Angela Di Capua
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro
2, 53100 Siena, Italy
- Griffith
Institute for Drug Discovery, Griffith University, Nathan 4111, Queensland, Australia
| | - Andrea Cappelli
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro
2, 53100 Siena, Italy
| | - Gianluca Giorgi
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro
2, 53100 Siena, Italy
| | - Mersedeh Farzad
- Oncological
Clinical Trials and Medical Oncology Unit, Alta Valdelsa Hospital, 53036 Campostaggia, Siena, Italy
| | - Lorenzo Di Cesare Mannelli
- Dipartimento
di Neuroscienze, Area del Farmaco e Salute del Bambino (NEUROFARBA), Viale Pieraccini, 6, 50139 Firenze, Italy
| | - Laura Micheli
- Dipartimento
di Neuroscienze, Area del Farmaco e Salute del Bambino (NEUROFARBA), Viale Pieraccini, 6, 50139 Firenze, Italy
| | - Carla Ghelardini
- Dipartimento
di Neuroscienze, Area del Farmaco e Salute del Bambino (NEUROFARBA), Viale Pieraccini, 6, 50139 Firenze, Italy
| | - Maurizio Anzini
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro
2, 53100 Siena, Italy
| |
Collapse
|
42
|
Chen X, Hu W, Lu X, Jiang B, Wang J, Zhang W, Huang C. Mechanism of 2,3,4',5-Tetrahydroxystilbene 2-O-β-D-Glucoside-Induced Upregulation of Glutamate Transporter 1 Protein Expression in Mouse Primary Astrocytes. Pharmacology 2017; 99:153-159. [PMID: 28049198 DOI: 10.1159/000452672] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/18/2016] [Indexed: 12/17/2022]
Abstract
Glutamate transporter-1 (GLT-1), a major glutamate transporter expressed in astrocytes, takes up excess glutamate from the micro-environment in order to prevent excitotoxicity. Drugs that increase GLT-1 expression may have therapeutic effects in disorders associated with neuronal excitotoxicity. 2,3,4',5-tetrahydroxystilbene 2-O-β-D-glucoside (TSG), a monomer of stilbene from polygonummultiflorum, exerts neuroprotection in a range of experimental models such as Alzheimer's disease and brain ischemia. In this study, we evaluated the effect of TSG on GLT-1 protein expression in mouse primary-cultured astrocytes. Results showed that TSG markedly increased the GLT-1 protein expression level in mouse primary-cultured astrocytes in a dose- and time-dependent manner, and this increase was mediated by the activation of protein kinase B (Akt) but not by the activation of extracellular signal-regulated protein kinase 1/2. Furthermore, inhibition of cAMP response element-binding protein, but not nuclear factor kappa B, abolished the TSG-mediated increase in GLT-1 protein expression in cultured astrocytes. Collectively, these findings may provide novel insights into the mechanism for TSG in neuroprotection, and would help search new agents targeting neurodegenerative disorders associated with impaired astrocytic glutamate transporters.
Collapse
Affiliation(s)
- Xiangfan Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Hefendehl JK, LeDue J, Ko RWY, Mahler J, Murphy TH, MacVicar BA. Mapping synaptic glutamate transporter dysfunction in vivo to regions surrounding Aβ plaques by iGluSnFR two-photon imaging. Nat Commun 2016; 7:13441. [PMID: 27834383 PMCID: PMC5114608 DOI: 10.1038/ncomms13441] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 10/04/2016] [Indexed: 12/22/2022] Open
Abstract
Amyloid-β (Aβ) plaques, a hallmark of Alzheimer's disease (AD), are surrounded by regions of neuronal and glial hyperactivity. We use in vivo two-photon and wide-field imaging of the glutamate sensor iGluSnFR to determine whether pathological changes in glutamate dynamics in the immediate vicinity of Aβ deposits in APPPS1 transgenic mice could alter neuronal activity in this microenvironment. In regions close to Aβ plaques chronic states of high spontaneous glutamate fluctuations are observed and the timing of glutamate responses evoked by sensory stimulation exhibit slower decay rates in two cortical brain areas. GLT-1 expression is reduced around Aβ plaques and upregulation of GLT-1 expression and activity by ceftriaxone partially restores glutamate dynamics to values in control regions. We conclude that the toxic microenvironment surrounding Aβ plaques results, at least partially, from enhanced glutamate levels and that pharmacologically increasing GLT-1 expression and activity may be a new target for early therapeutic intervention. In Alzheimer's disease (AD), neural hyperactivity has been shown to occur in the regions surrounding Aβ plaques. Here, the authors use in vivo two-photon imaging in mouse models of AD and report abnormal glutamate dynamics in the vicinity of plaques which can be partially restored via GLT-1 upregulation through Ceftriaxone treatment.
Collapse
Affiliation(s)
- J K Hefendehl
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - J LeDue
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - R W Y Ko
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - J Mahler
- Hertie-Institut für klinische Hirnforschung, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - T H Murphy
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - B A MacVicar
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, Canada V6T 1Z3
| |
Collapse
|
44
|
Clonidine preconditioning alleviated focal cerebral ischemic insult in rats via up-regulating p-NMDAR1 and down-regulating NMDAR2A / p-NMDAR2B. Eur J Pharmacol 2016; 793:89-94. [PMID: 27806917 DOI: 10.1016/j.ejphar.2016.10.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 10/22/2016] [Accepted: 10/24/2016] [Indexed: 01/17/2023]
Abstract
A brain ischemia rat model was established by middle cerebral artery occlusion (MCAO) for 2h and reperfusion for 4h to investigate the underlying mechanism of the neuroprotection action of clonidine, a classical alpha-2 adrenergic agonist, on cerebral ischemia. Clonidine and yohimbine were intraperitoneally given to the rats each day for a week before ischemia. Neurological deficits evaluations were carried out at 6h after operation. TTC staining method was used to measure the volume of brain infarction. Expression levels of NMDAR1, NMDAR2A, NMDAR2B were assayed by western blotting. Our data demonstrated that clonidine pretreatment significantly improved the neurological deficit scores and reduced the brain infarct volumes of the rats. Furthermore, protein expression level of p-NMDAR2B in cortex was significantly up-regulated whereas that of p-NMDAR1 was decreased when compared with the sham-operated rats. Remarkably, clonidine treatment led to significant down-regulation of p-NMDAR2B and NMDAR2A in addition to enhancement of the expression level of p-NMDAR1 in cortex. This is the first report illustrating the neuroprotective role of clonidine may be mediated through modulation of the expression levels of p-NMDAR2B, NMDAR2A and p-NMDAR1 during cerebral ischemia.
Collapse
|
45
|
Gong HY, Zheng F, Zhang C, Chen XY, Liu JJ, Yue XQ. Propofol protects hippocampal neurons from apoptosis in ischemic brain injury by increasing GLT-1 expression and inhibiting the activation of NMDAR via the JNK/Akt signaling pathway. Int J Mol Med 2016; 38:943-50. [DOI: 10.3892/ijmm.2016.2663] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 06/21/2016] [Indexed: 11/06/2022] Open
|
46
|
Wang JG, Wang YL, Xu F, Zhao JX, Zhou SY, Yu Y, Chazot PL, Wang XF, Lu CB. Activity- and development-dependent down-regulation of TARPγ8 and GluA1 in cultured rat hippocampal neurons. Acta Pharmacol Sin 2016; 37:303-11. [PMID: 26725511 DOI: 10.1038/aps.2015.112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/26/2015] [Indexed: 01/16/2023] Open
Abstract
AIM Transmembrane AMPA receptor regulatory proteins (TARPs) regulate the trafficking and expression of AMPA receptors that are essential for the fast excitatory synaptic transmission and plasticity in the brain. This study aimed to investigate the activity-dependent regulation of TARPγ8 in cultured rat hippocampal neurons. METHODS Rat hippocampal neurons cultured for 7-8 DIV or 17-18 DIV were exposed to the AMPA receptor agonist AMPA at a non-toxic concentration (100 μmol/L) for 4 h. The protein levels of TARPγ8 and AMPA receptor subunits (GluA1 and GluA2) were measured using Western blotting analysis. AMPA-induced currents were recorded in the neurons using a whole-cell recording method. RESULTS Four-hour exposure to AMPA significantly decreased the protein levels of TARPγ8 and GluA1 in the neurons at 17-18 DIV, but did not change the protein level of TARPγ8 in the neurons cultured at 7-8 DIV. AMPA-induced down-regulation of TARPγ8 and GluA1 was largely blocked by the calpain inhibitor calpeptin (50 μmol/L), but not affected by the caspase inhibitor zVAD (50 μmol/L). Four-hour exposure to AMPA significantly decreased AMPA-induced currents in the neurons at 17-18 DIV, which was blocked by co-exposure to calpeptin (50 μmol/L). CONCLUSION The down-regulation of TARPγ8 and GluA1 protein levels and AMPA-induced currents in cultured rat hippocampal neurons is activity- and development-dependent, and mediated by endogenous calpain.
Collapse
|
47
|
Lee KE, Cho KO, Choi YS, Kim SY. The neuroprotective mechanism of ampicillin in a mouse model of transient forebrain ischemia. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:185-92. [PMID: 26937215 PMCID: PMC4770109 DOI: 10.4196/kjpp.2016.20.2.185] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 01/16/2016] [Accepted: 01/23/2016] [Indexed: 11/22/2022]
Abstract
Ampicillin, a β-lactam antibiotic, dose-dependently protects neurons against ischemic brain injury. The present study was performed to investigate the neuroprotective mechanism of ampicillin in a mouse model of transient global forebrain ischemia. Male C57BL/6 mice were anesthetized with halothane and subjected to bilateral common carotid artery occlusion for 40 min. Before transient forebrain ischemia, ampicillin (200 mg/kg, intraperitoneally [i.p.]) or penicillin G (6,000 U/kg or 20,000 U/kg, i.p.) was administered daily for 5 days. The pretreatment with ampicillin but not with penicillin G signifi cantly attenuated neuronal damage in the hippocampal CA1 subfield. Mechanistically, the increased activity of matrix metalloproteinases (MMPs) following forebrain ischemia was also attenuated by ampicillin treatment. In addition, the ampicillin treatment reversed increased immunoreactivities to glial fibrillary acidic protein and isolectin B4, markers of astrocytes and microglia, respectively. Furthermore, the ampicillin treatment significantly increased the level of glutamate transporter-1, and dihydrokainic acid (DHK, 10 mg/kg, i.p.), an inhibitor of glutamate transporter-1 (GLT-1), reversed the neuroprotective effect of ampicillin. Taken together, these data indicate that ampicillin provides neuroprotection against ischemia-reperfusion brain injury, possibly through inducing the GLT-1 protein and inhibiting the activity of MMP in the mouse hippocampus.
Collapse
Affiliation(s)
- Kyung-Eon Lee
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Kyung-Ok Cho
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Yun-Sik Choi
- Department of Pharmaceutical Science and Technology, College of Health and Medical Science, Catholic University of Daegu, Daegu 712-702, Korea
| | - Seong Yun Kim
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| |
Collapse
|
48
|
Takahashi K, Foster JB, Lin CLG. Glutamate transporter EAAT2: regulation, function, and potential as a therapeutic target for neurological and psychiatric disease. Cell Mol Life Sci 2015; 72:3489-506. [PMID: 26033496 PMCID: PMC11113985 DOI: 10.1007/s00018-015-1937-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 12/12/2022]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the central nervous system. Excitatory amino acid transporter 2 (EAAT2) is primarily responsible for clearance of extracellular glutamate to prevent neuronal excitotoxicity and hyperexcitability. EAAT2 plays a critical role in regulation of synaptic activity and plasticity. In addition, EAAT2 has been implicated in the pathogenesis of many central nervous system disorders. In this review, we summarize current understanding of EAAT2, including structure, pharmacology, physiology, and functions, as well as disease relevancy, such as in stroke, Parkinson's disease, epilepsy, amyotrophic lateral sclerosis, Alzheimer's disease, major depressive disorder, and addiction. A large number of studies have demonstrated that up-regulation of EAAT2 protein provides significant beneficial effects in many disease models suggesting EAAT2 activation is a promising therapeutic approach. Several EAAT2 activators have been identified. Further understanding of EAAT2 regulatory mechanisms could improve development of drug-like compounds that spatiotemporally regulate EAAT2.
Collapse
Affiliation(s)
- Kou Takahashi
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Joshua B. Foster
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Chien-Liang Glenn Lin
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| |
Collapse
|
49
|
Fontana ACK. Current approaches to enhance glutamate transporter function and expression. J Neurochem 2015; 134:982-1007. [DOI: 10.1111/jnc.13200] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Andréia C. K. Fontana
- Department of Pharmacology and Physiology; Drexel University College of Medicine; Philadelphia Pennsylvania USA
| |
Collapse
|
50
|
Tong H, Yu X, Lu X, Wang P. Downregulation of solute carriers of glutamate in gliosomes and synaptosomes may explain local brain metastasis in anaplastic glioblastoma. IUBMB Life 2015; 67:306-11. [PMID: 25914026 DOI: 10.1002/iub.1372] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 02/28/2015] [Indexed: 12/31/2022]
Abstract
Advanced grades of glioblastoma are highly aggressive, especially in terms of multisite spread within the brain or even to distant sites at the spinal cord. In advanced grades of glioblastoma, glutamate and glutamine are reported to be increased in concentration in the extracellular fluid. It has been reported that glutamate acts as an extracellular signaling molecule for facilitating local spread of advanced grades of glioblastoma. In the present study, we aimed to examine whether glutamate uptake mechanisms is impaired in advanced glioblastoma. The possible downregulated mechanisms of glutamate uptake would facilitate persistence of glutamate in the extracellular environment, rather than intracellular uptake. We obtained biobanked human specimens of glioblastoma and tested expression of proteins belonging to the solute carrier families of proteins that are known to function as membrane-located excitatory amino acid like glutamate transporters. The present study provides preliminary evidence of the downregulation of membrane expression of excitatory amino acid transporters solute carrier family 1 member 3 (SLC1A3) and its palmitoylated form in gliosomes, as well as SLC1A2 in the glio-synaptosomes. Compounds like riluzole used in the treatment of amyotrophic lateral sclerosis and the antibiotic ceftriaxone have the potential to facilitate glutamate uptake. These medications may be examined as adjunct chemotherapy in the massively aggressive tumor glioblastoma multiforme.
Collapse
Affiliation(s)
- Huaiyu Tong
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xinguang Yu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xuechun Lu
- Department of Hematology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Peng Wang
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|