1
|
Bath PM, Howard G, Hacke W. The hazards of chasing subgroups in neutral stroke trials. Neurol Res Pract 2025; 7:17. [PMID: 40069910 PMCID: PMC11921980 DOI: 10.1186/s42466-025-00369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/13/2025] [Indexed: 03/21/2025] Open
Abstract
BACKGROUND The majority of randomised controlled trials in acute stroke and many for prevention are neutral, i.e. they failed to reach statistical significance. However, many of these will find apparent benefit in a component of a subgroup, findings which may be 'chased' in a follow-up trial. The evidence to date is that these follow-on trials are very likely to be neutral. FINDINGS We discuss the issue of chasing subgroups in neutral trials and illustrate the challenges in five pairs of exemplar acute stroke trials. Problems in the exemplar trials include failing to define the subgroup in advance or even changing its definition, failing to show that both the interaction test and the primary outcome in the component were statistically significant, failing to publish additional information on the positive subgroup component, having too many subgroups, failing to make the follow-on trial large enough and failing to report the findings of the follow-on trial. CONCLUSION When chasing a positive component in a subgroup, it is vital that the subgroup: should be plausible biologically, defined a priori and have a significant interaction test. Further the number of subgroups should be limited and the component of interest should be statistically significant. Explanations should be given as to why the component is positive and other components of the subgroup are negative. Other outcomes should also show potential benefit. Unless this guidance is followed, it is highly likely that follow-on trials will be neutral as has occurred previously.
Collapse
Affiliation(s)
- Philip M Bath
- Stroke Trials Unit, Mental Health & Clinical Neuroscience, Queens Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK.
| | - George Howard
- School of Public Health - Biostatistics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Werner Hacke
- Department of Neurology, Ruprechts Karl University Heidelberg, Heidelberg, Germany
| |
Collapse
|
2
|
Perovic M, Pavlovic D, Palmer Z, Udo MSB, Citadin CT, Rodgers KM, Wu CYC, Zhang Q, Lin HW, Tesic V. Modulation of GABAergic system as a therapeutic option in stroke. Exp Neurol 2025; 384:115050. [PMID: 39522803 DOI: 10.1016/j.expneurol.2024.115050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/18/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Stroke is one of the leading causes of death and permanent adult disability worldwide. Despite the improvements in reducing the rate and mortality, the societal burden and costs of treatment associated with stroke management are increasing. Most of the therapeutic approaches directly targeting ischemic injury have failed to reduce short- and long-term morbidity and mortality and more effective therapeutic strategies are still needed to promote post-stroke functional recovery. Decades of stroke research have been focused on hyperexcitability and glutamate-induced excitotoxicity in the acute phase of ischemia and their relation to motor deficits. Recent advances in understanding the pathophysiology of stroke have been made with several lines of evidence suggesting that changes in the neurotransmission of the major inhibitory system via γ-Aminobutyric acid (GABA) play a particularly important role in functional recovery and deserve further attention. The present review provides an overview of how GABAergic neurotransmission changes correlate with stroke recovery and outlines GABAergic system modulators with special emphasis on neurosteroids that have been shown to affect stroke pathogenesis or plasticity or to protect against cognitive decline. Supporting evidence from both animal and human clinical studies is presented and the potential for GABA signaling-targeted therapies for stroke is discussed to translate this concept to human neural repair therapies. Age and sex are considered crucial parameters related to the pathophysiology of stroke and important factors in the development of therapeutic pharmacological strategies. Future work is needed to deepen our knowledge of the neurochemical changes after stroke, extend the conceptual framework, and allow for the development of more effective interventions that include the modulation of the inhibitory system.
Collapse
Affiliation(s)
- Milka Perovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Damjan Pavlovic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Zoe Palmer
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Mariana S B Udo
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Cristiane T Citadin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Krista M Rodgers
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Celeste Yin-Chien Wu
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| |
Collapse
|
3
|
Mordyl B, Fajkis-Zajączkowska N, Szafrańska K, Siwek A, Głuch-Lutwin M, Żmudzki P, Jończyk J, Karcz T, Słoczyńska K, Pękala E, Pomierny B, Krzyżanowska W, Jurczyk J, Skórkowska A, Sałach A, Jastrzębska-Więsek M, Walczak M, Gawlik MT, Smolik M, Kolaczkowski M, Marcinkowska M. Preferential Synaptic Type of GABA-A Receptor Ligands Enhancing Neuronal Survival and Facilitating Functional Recovery After Ischemic Stroke. J Med Chem 2024; 67:21859-21889. [PMID: 39668673 DOI: 10.1021/acs.jmedchem.4c01578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Selective enhancement of synaptic GABA signaling mediated by GABA-A receptors has been previously reported to promote functional recovery after ischemic stroke, while tonic GABA signaling has been detrimental. To identify agents that enhance synaptic signaling, we synthesized GABA-A ligands based on three chemotypes with affinity values pKi= 6.44-8.32. Representative compounds showed a preference in functional responses toward synaptic type of GABA-A receptors, compared to the extrasynaptic ones. In a cellular ischemia model (OGD), selected compounds showed the potential to improve neuronal recovery. The selected lead, compound 4, demonstrated the ability to reduce mitochondrial dysfunction, regulate intracellular calcium levels, decrease caspase 3 levels, and promote neurite outgrowth in in vitro assays. In an animal model, compound 4 enhanced motor recovery and showed neuroprotective activity by reducing infarct volume and decreasing poststroke acidosis. These findings underscore the value of selective ligands modulating synaptic GABA-A receptors in promoting recovery from ischemic stroke.
Collapse
Affiliation(s)
- Barbara Mordyl
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Nikola Fajkis-Zajączkowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Katarzyna Szafrańska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., Krakow 31-530, Poland
| | - Agata Siwek
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
- Center for the Development of Therapies for Civilization and Age-Related Diseases, Jagiellonian University Medical College, Skawińska 8, Krakow 31-066, Poland
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Paweł Żmudzki
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
- Center for the Development of Therapies for Civilization and Age-Related Diseases, Jagiellonian University Medical College, Skawińska 8, Krakow 31-066, Poland
| | - Jakub Jończyk
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Karolina Słoczyńska
- Department of Pharmaceutical Biochemistry, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Bartosz Pomierny
- Center for the Development of Therapies for Civilization and Age-Related Diseases, Jagiellonian University Medical College, Skawińska 8, Krakow 31-066, Poland
- Department of Toxicological Biochemistry, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Weronika Krzyżanowska
- Department of Toxicological Biochemistry, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Jakub Jurczyk
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., Krakow 31-530, Poland
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Alicja Skórkowska
- Center for the Development of Therapies for Civilization and Age-Related Diseases, Jagiellonian University Medical College, Skawińska 8, Krakow 31-066, Poland
- Department of Toxicological Biochemistry, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Aleksandra Sałach
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Magdalena Jastrzębska-Więsek
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Maria Walczak
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Maciej Tadeusz Gawlik
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Magdalena Smolik
- Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Marcin Kolaczkowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Monika Marcinkowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
- Center for the Development of Therapies for Civilization and Age-Related Diseases, Jagiellonian University Medical College, Skawińska 8, Krakow 31-066, Poland
| |
Collapse
|
4
|
Washabaugh EP, Foley SA, Czopek EG, Krishnan C. Altered Corticospinal and Intracortical Excitability After Stroke: A Systematic Review With Meta-Analysis. Neurorehabil Neural Repair 2024; 38:845-862. [PMID: 39275953 DOI: 10.1177/15459683241281299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
BACKGROUND Intracortical inhibitory/faciliatory measures are affected after stroke; however, the evidence is conflicting. OBJECTIVE This meta-analysis aimed to investigate the changes in motor threshold (MT), motor evoked potential (MEP), short-interval intracortical inhibition (SICI), and intracortical facilitation (ICF), and identify sources of study variability using a machine learning approach. METHODS We identified studies that objectively evaluated corticospinal excitability and intracortical inhibition/facilitation after stroke using transcranial magnetic stimulation. Pooled within- (ie, affected hemisphere [AH] vs unaffected hemisphere [UH]) and between-subjects (ie, AH and UH vs Control) standardized mean differences were computed. Decision trees determined which factors accurately predicted studies that showed alterations in corticospinal excitability and intracortical inhibition/facilitation. RESULTS A total of 35 studies (625 stroke patients and 328 healthy controls) were included. MT was significantly increased and MEP was significantly decreased (ie, reduced excitability) in the AH when compared with the UH and Control (P < .01). SICI was increased (ie, reduced inhibition) for the AH when compared with the UH, and for the AH and UH when compared with Control (P < .001). ICF was significantly increased (ie, increased facilitation) in the AH when compared with UH (P = .016) and decreased in UH when compared with Control (P < 0.001). Decision trees indicated that demographic and methodological factors accurately predicted (73%-86%) studies that showed alterations in corticospinal and intracortical excitability measures. CONCLUSIONS The findings indicate that stroke alters corticospinal and intracortical excitability measures. Alterations in SICI and ICF may reflect disinhibition of the motor cortex after stroke, which is contrary to the notion that stroke increases inhibition of the affected side.
Collapse
Affiliation(s)
- Edward P Washabaugh
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - Sierra A Foley
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - Emily G Czopek
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - Chandramouli Krishnan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Physical Medicine and Rehabilitation, Michigan Medicine, Ann Arbor, MI, USA
- Robotics Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Physical Therapy Department, University of Michigan-Flint, Flint, MI, USA
| |
Collapse
|
5
|
Heit BS, Chu A, McRay A, Richmond JE, Heckman CJ, Larson J. Interference with glutamate antiporter system x c - enables post-hypoxic long-term potentiation in hippocampus. Exp Physiol 2024; 109:1572-1592. [PMID: 39153228 PMCID: PMC11363115 DOI: 10.1113/ep092045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 08/19/2024]
Abstract
Our group previously showed that genetic or pharmacological inhibition of the cystine/glutamate antiporter, system xc -, mitigates excitotoxicity after anoxia by increasing latency to anoxic depolarization, thus attenuating the ischaemic core. Hypoxia, however, which prevails in the ischaemic penumbra, is a condition where neurotransmission is altered, but excitotoxicity is not triggered. The present study employed mild hypoxia to further probe ischaemia-induced changes in neuronal responsiveness from wild-type and xCT KO (xCT-/-) mice. Synaptic transmission was monitored in hippocampal slices from both genotypes before, during and after a hypoxic episode. Although wild-type and xCT-/- slices showed equal suppression of synaptic transmission during hypoxia, mutant slices exhibited a persistent potentiation upon re-oxygenation, an effect we termed 'post-hypoxic long-term potentiation (LTP)'. Blocking synaptic suppression during hypoxia by antagonizing adenosine A1 receptors did not preclude post-hypoxic LTP. Further examination of the induction and expression mechanisms of this plasticity revealed that post-hypoxic LTP was driven by NMDA receptor activation, as well as increased calcium influx, with no change in paired-pulse facilitation. Hence, the observed phenomenon engaged similar mechanisms as classical LTP. This was a remarkable finding as theta-burst stimulation-induced LTP was equivalent between genotypes. Importantly, post-hypoxic LTP was generated in wild-type slices pretreated with system xc - inhibitor, S-4-carboxyphenylglycine, thereby confirming the antiporter's role in this phenomenon. Collectively, these data indicate that system xc - interference enables neuroplasticity in response to mild hypoxia, and, together with its regulation of cellular damage in the ischaemic core, suggest a role for the antiporter in post-ischaemic recovery of the penumbra.
Collapse
Affiliation(s)
- Bradley S. Heit
- Department of Neuroscience and Department of Biomedical EngineeringNorthwestern UniversityChicagoIllinoisUSA
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alex Chu
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Alyssa McRay
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Janet E. Richmond
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Charles J. Heckman
- Department of Neuroscience and Department of Biomedical EngineeringNorthwestern UniversityChicagoIllinoisUSA
| | - John Larson
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of Biological SciencesUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
6
|
Cui Y, Cui M, Wang L, Wang N, Chen Y, Lv S, Zhang L, Chen C, Yang Y, Wang F, Wang L, Cui H. Huanglian Jiedu decoction alleviates ischemia-induced cerebral injury in rats by mitigating NET formation and activiting GABAergic synapses. J Cell Mol Med 2024; 28:e18528. [PMID: 39099086 PMCID: PMC11298410 DOI: 10.1111/jcmm.18528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 08/06/2024] Open
Abstract
Huanglian Jiedu decoction (HLJD) has been used to treat ischemic stroke in clinic. However, the detailed protective mechanisms of HLJD on ischemic stroke have yet to be elucidated. The aim of this study is to elucidate the underlying pharmacological mechanisms of HLJD based on the inhibition of neuroinflammation and the amelioration of nerve cell damage. A middle cerebral artery occlusion reperfusion (MCAO/R) model was established in rats and received HLJD treatment. Effects of HLJD on neurological function was assessed based on Bederson's score, postural reflex test and asymmetry score. 2, 3, 5-Triphenyltetrazolium chloride (TTC) staining, Hematein and eosin (HE) and Nissl staining were used to observe the pathological changes in brain. Then, transcriptomics was used to screen the differential genes in brain tissue in MCAO/R model rats following HLJD intervention. Subsequently, the effects of HLJD on neutrophil extracellular trap (NET) formation-related neuroinflammation, gamma-aminobutyric acid (GABA)ergic synapse activation, nerve cell damage and proliferation were validated using immunofluorescence, western blot and enzyme-linked immunosorbent assay (ELISA). Our results showed that HLJD intervention reduced the Bederson's score, postural reflex test score and asymmetry score in MCAO/R model rats. Pathological staining indicated that HLJD treatment decreased the cerebral infarction area, mitigated neuronal damage and increased the numbers of Nissl bodies. Transcriptomics suggested that HLJD affected 435 genes in MCAO/R rats. Among them, several genes involving in NET formation and GABAergic synapses pathways were dysregulated. Subsequent experimental validation showed that HLJD reduced the MPO+CitH3+ positive expression area, reduced the protein expression of PAD4, p-P38/P38, p-ERK/ERK and decreased the levels of IL-1β, IL-6 and TNF-α, reversed the increase of Iba1+TLR4+, Iba1+p65+ and Iba1+NLRP3+ positive expression area in brain. Moreover, HLJD increased GABA levels, elevated the protein expression of GABRG1 and GAT3, decreased the TUNEL positive expression area and increased the Ki67 positive expression area in brain. HLJD intervention exerts a multifaceted positive impact on ischemia-induced cerebral injury in MCAO/R rats. This intervention effectively inhibits neuroinflammation by mitigating NET formation, and concurrently improves nerve cell damage and fosters nerve cell proliferation through activating GABAergic synapses.
Collapse
Affiliation(s)
- Youxiang Cui
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Mingyue Cui
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Leilei Wang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Ning Wang
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Yao Chen
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Shuquan Lv
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Limin Zhang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Congai Chen
- Beijing University of Chinese MedicineBeijingChina
| | - Yanwen Yang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Feng Wang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Lichun Wang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Huantian Cui
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| |
Collapse
|
7
|
Lan J, Wang J, Wang S, Wang J, Huang S, Wang Y, Ma Y. The Activation of GABA AR Alleviated Cerebral Ischemic Injury via the Suppression of Oxidative Stress, Autophagy, and Apoptosis Pathways. Antioxidants (Basel) 2024; 13:194. [PMID: 38397792 PMCID: PMC10886019 DOI: 10.3390/antiox13020194] [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: 12/12/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Ischemic stroke is a devastating disease leading to neurologic impairment. Compounding the issue is the very limited array of available interventions. The activation of a γ-aminobutyric acid (GABA) type A receptor (GABAAR) has been reported to produce neuroprotective properties during cerebral ischemia, but its mechanism of action is not yet fully understood. Here, in a rat model of photochemically induced cerebral ischemia, we found that muscimol, a GABAAR agonist, modulated GABAergic signaling, ameliorated anxiety-like behaviors, and attenuated neuronal damage in rats suffering cerebral ischemia. Moreover, GABAAR activation improved brain antioxidant levels, reducing the accumulation of oxidative products, which was closely associated with the NO/NOS pathway. Notably, the inhibition of autophagy markedly relieved the neuronal insult caused by cerebral ischemia. We further established an oxygen-glucose deprivation (OGD)-induced PC12 cell injury model. Both in vivo and in vitro experiments demonstrated that GABAAR activation obviously suppressed autophagy by regulating the AMPK-mTOR pathway. Additionally, GABAAR activation inhibited apoptosis through inhibiting the Bax/Bcl-2 pathway. These data suggest that GABAAR activation exerts neuroprotective effects during cerebral ischemia through improving oxidative stress and inhibiting autophagy and apoptosis. Our findings indicate that GABAAR serves as a target for treating cerebral ischemia and highlight the GABAAR-mediated autophagy signaling pathway.
Collapse
Affiliation(s)
- Jing Lan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiaqi Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shujing Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jia Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Sijuan Huang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, The Fourth Military Medical University, Xi’an 710032, China
| | - Yunfei Ma
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
8
|
Danilov AB, Shindryaeva NN, Borodulina IV, Lunegov TD. [Integrative assessment of the effectiveness and safety of outpatient use of Picamilon]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:119-130. [PMID: 39113452 DOI: 10.17116/jnevro2024124071119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
OBJECTIVE To study the efficacy and safety of the use of the drug Picamilon with various therapy regimens in patients with stage I of chronic cerebral ischemia (CCI). MATERIAL AND METHODS Material and methods. An open randomized comparative clinical trial included 44 patients with stage CCI aged 46 to 67 years (average age 55.6±6.76 years). Patients were randomized into two groups, patients of group 1 (n=23) received Picamilon orally in tablets of 50 mg 3 times/day for 60 days; group 2 (n=21) received Picamilon first parenterally at 100 mg i/m for 10 days, then in tablets of 50 mg 3 times/day for 50 days. The total duration of therapy was 60 days. The study included 4 visits (before treatment, 10 days later, 60 days later, 1.5 months after completion of treatment). The dynamics of cognitive status were assessed according to the Montreal Cognitive Function Assessment Scale (MoCA), vegetative disorders on the A.M. Wayne scale, neurological disorders on the A.I. Fedin scale, and sleep quality on the Ya.I. Levin scale. The study of the state of cerebral blood flow (dopplerography of intracranial vessels) and endothelial function (assessment of the level of methylation. RESULTS During treatment, in the total sample of patients, there was a positive trend in the results of the MoCA scale, increasing in the delayed period (24.9/26.5/28.3 points, p=0.022 and p<0.001); improvement in sleep quality in 50% of patients by visit 3 and in 84% by visit 4, in the 2nd group the effect occurred in 28% of patients, in the 1st - in 11%, by the end of the study the effect was comparable (p=0.508). Improvement according to Fedin A.I. scale noted in 77% of patients, values decreased from 11.9±8.3 to 6±6.1 points (p<0.0001) and to 2.77±4.43 points by visit 4 (p<0.0001). Normalization of autonomic functions was observed in 29% of patients (p=0.024) without intergroup differences. Picamilon therapy showed high efficacy in terms of clinical outcomes (up to 89%), good tolerability (98% of patients) and a favorable safety profile (less than 8.6% of AEs). The use of Picamilon was accompanied by an increase in the linear velocity of blood flow, a decrease in the thickness of the intima-media complex and the resistance index; a decrease in elevated ADMA concentrations and ADMA/MMA and (ADMA+SDMA)/MMA ratios. CONCLUSION The use of Picamilon is effective in patients with stage I CCI, contributes to a significant regression of neurological deficits, cognitive impairment, improved sleep quality and autonomic function; improves vascular endothelial function, reduces the risk of atherosclerosis and cardiovascular complications in patients. The optimal duration of therapy with Picamilon in stage I of chronic cerebral ischemia is 2 months.
Collapse
Affiliation(s)
- A B Danilov
- Sechenov First Moscow State Medical University (Sechenovsky University), Moscow, Russia
| | - N N Shindryaeva
- Sechenov First Moscow State Medical University (Sechenovsky University), Moscow, Russia
| | - I V Borodulina
- Russian Medical Academy of Continuing Postgraduate Education, Moscow, Russia
| | | |
Collapse
|
9
|
Liu CH, Nguyen HTM, Lee DY, Hsieh CL. Effect of electrode configuration in electroacupuncture on ischemic stroke treatment in rats. J Tradit Complement Med 2023; 13:588-599. [PMID: 38020550 PMCID: PMC10658373 DOI: 10.1016/j.jtcme.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/22/2023] [Accepted: 09/15/2023] [Indexed: 12/01/2023] Open
Abstract
Background and aim This study investigated the effect of the electrode configuration on EA treating ischemic stroke. Experimental procedure An ischemic stroke rat model was established. In the EA-P group, the anodes of EA were placed on the BL7 and BL8 acupoints of the lesioned, and the cathodes were placed on the BL7 and BL8 acupoints of the nonlesioned hemispheres; by contrast, in the EA-N group. Results The difference in neurological deficit scores between the first and fourth days and the difference in Rotarod test time between the fourth and first days after reperfusion were greater in the EA-P and EA-N groups than in the sham group (all p < 0.001). In the lesioned hemisphere, neuronal nuclei (NeuN), γ-aminobutyric acid-A (GABA)-A, postsynaptic density 95 (PSD95), and astrocyte glutamate transporter 1 (GLT-1) expression and microtubule-associated protein 2 (MAP2)/glyceraldehyde 3-phosphate dehydrogenase (GADPH) ratios were greater and the glial fibrillary acid protein (GFAP)/GADPH ratios were smaller in the EA-P than in the sham group (all p < 0.05), but these ratios in the EA-N group were similar to those in the sham group (all p > 0.05); serum adrenaline and serotonin levels in the sham group were lower than those in the normal and EA-P groups (both p < 0.05), and cerebrospinal fluid (CSF) glutamate levels were higher in the EA-P group than in the sham group (p < 0.05). Conclusion EA improved neurological function through multiple pathways. However, placing the anode on the lesioned hemisphere can provide more neuroprotection.
Collapse
Affiliation(s)
- Chung-Hsiang Liu
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Huong Thi Mai Nguyen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Der-Yen Lee
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Liang Hsieh
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
10
|
Zhang T, Deng D, Huang S, Fu D, Wang T, Xu F, Ma L, Ding Y, Wang K, Wang Y, Zhao W, Chen X. A retrospect and outlook on the neuroprotective effects of anesthetics in the era of endovascular therapy. Front Neurosci 2023; 17:1140275. [PMID: 37056305 PMCID: PMC10086253 DOI: 10.3389/fnins.2023.1140275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Studies on the neuroprotective effects of anesthetics were carried out more than half a century ago. Subsequently, many cell and animal experiments attempted to verify the findings. However, in clinical trials, the neuroprotective effects of anesthetics were not observed. These contradictory results suggest a mismatch between basic research and clinical trials. The Stroke Therapy Academic Industry Roundtable X (STAIR) proposed that the emergence of endovascular thrombectomy (EVT) would provide a proper platform to verify the neuroprotective effects of anesthetics because the haemodynamics of patients undergoing EVT is very close to the ischaemia–reperfusion model in basic research. With the widespread use of EVT, it is necessary for us to re-examine the neuroprotective effects of anesthetics to guide the use of anesthetics during EVT because the choice of anesthesia is still based on team experience without definite guidelines. In this paper, we describe the research status of anesthesia in EVT and summarize the neuroprotective mechanisms of some anesthetics. Then, we focus on the contradictory results between clinical trials and basic research and discuss the causes. Finally, we provide an outlook on the neuroprotective effects of anesthetics in the era of endovascular therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiangdong Chen
- *Correspondence: Xiangdong Chen, ; orcid.org/0000-0003-3347-2947
| |
Collapse
|
11
|
Selective ischemic-hemisphere targeting Ginkgolide B liposomes with improved solubility and therapeutic efficacy for cerebral ischemia-reperfusion injury. Asian J Pharm Sci 2023; 18:100783. [PMID: 36891470 PMCID: PMC9986716 DOI: 10.1016/j.ajps.2023.100783] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/23/2022] [Accepted: 01/24/2023] [Indexed: 02/13/2023] Open
Abstract
Cerebral ischemia-reperfusion injury (CI/RI) remains the main cause of disability and death in stroke patients due to lack of effective therapeutic strategies. One of the main issues related to CI/RI treatment is the presence of the blood-brain barrier (BBB), which affects the intracerebral delivery of drugs. Ginkgolide B (GB), a major bioactive component in commercially available products of Ginkgo biloba, has been shown significance in CI/RI treatment by regulating inflammatory pathways, oxidative damage, and metabolic disturbance, and seems to be a candidate for stroke recovery. However, limited by its poor hydrophilicity and lipophilicity, the development of GB preparations with good solubility, stability, and the ability to cross the BBB remains a challenge. Herein, we propose a combinatorial strategy by conjugating GB with highly lipophilic docosahexaenoic acid (DHA) to obtain a covalent complex GB-DHA, which can not only enhance the pharmacological effect of GB, but can also be encapsulated in liposomes stably. The amount of finally constructed Lipo@GB-DHA targeting to ischemic hemisphere was validated 2.2 times that of free solution in middle cerebral artery occlusion (MCAO) rats. Compared to the marketed ginkgolide injection, Lipo@GB-DHA significantly reduced infarct volume with better neurobehavioral recovery in MCAO rats after being intravenously administered both at 2 h and 6 h post-reperfusion. Low levels of reactive oxygen species (ROS) and high neuron survival in vitro was maintained via Lipo@GB-DHA treatment, while microglia in the ischemic brain were polarized from the pro-inflammatory M1 phenotype to the tissue-repairing M2 phenotype, which modulate neuroinflammatory and angiogenesis. In addition, Lipo@GB-DHA inhibited neuronal apoptosis via regulating the apoptotic pathway and maintained homeostasis by activating the autophagy pathway. Thus, transforming GB into a lipophilic complex and loading it into liposomes provides a promising nanomedicine strategy with excellent CI/RI therapeutic efficacy and industrialization prospects.
Collapse
|
12
|
JM-20 affects GABA neurotransmission in Caenorhabditis elegans. Neurotoxicology 2022; 93:37-44. [PMID: 36029931 DOI: 10.1016/j.neuro.2022.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/22/2022]
Abstract
Along with the discovery of new candidate molecules for pharmaceuticals, several studies have emerged showing different mechanisms of action and toxicological aspects. 3-ethoxycarbonyl-2-methyl-4- (2-nitrophenyl)4,11-dihydro-1H-pyrido [2,3-b] [1,5] benzodiazepine (JM-20) is a hybrid molecule. It is derived from 1,5-benzodiazepines and structurally differentiated by the addition of 1,4-dihydropyridine bonded to the benzodiazepine ring. This gives this molecule potential neuroprotective, antioxidant, and anxiolytic activity. As this is a promising multi-target molecule, further studies are necessary to improve the knowledge about its mechanism of action. In our study, we used Caenorhabditis elegans (C. elegans) to investigate the effects of chronic treatment with JM-20. Nematodes from the wild-type strain (N2) were treated chronically at different concentrations of JM-20. Our results show that JM-20 does not cause mortality, but higher concentrations can delay the development of worms after 48h exposure. We assessed basic behaviors in the worm, and our data demonstrate decreased defecation cycle. Our results suggest that JM-20 acts on the C. elegans GABAergic system because GABA neurotransmission is associated with the worm intestine. We also observed increased locomotor activity and decreased egg-laying after JM-20 treatment. When both behaviors were evaluated in mutants with have reduced levels of GABA (unc-25), this effect is no observed, suggesting the GABAergic modulation. Still, the JM-20 exert similar effect of Diazepam in basic behaviors observed. To reinforce neuromodulatory action, computational analysis was performed, and results showed a JM-20 binding on allosteric sites of nematodes GABA receptors. Overall, this work provided a better understanding of the effects of JM-20 in C. elegans as well as showed the effects of this new molecule on the GABAergic system in this animal model.
Collapse
|
13
|
Gonzales NR, Grotta JC. Pharmacologic Modification of Acute Cerebral Ischemia. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
14
|
Liew SL, Lin DJ, Cramer SC. Interventions to Improve Recovery After Stroke. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00061-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
15
|
Lamtahri R, Hazime M, Gowing EK, Nagaraja RY, Maucotel J, Alasoadura M, Quilichini PP, Lehongre K, Lefranc B, Gach-Janczak K, Marcher AB, Mandrup S, Vaudry D, Clarkson AN, Leprince J, Chuquet J. The Gliopeptide ODN, a Ligand for the Benzodiazepine Site of GABA A Receptors, Boosts Functional Recovery after Stroke. J Neurosci 2021; 41:7148-7159. [PMID: 34210784 PMCID: PMC8372017 DOI: 10.1523/jneurosci.2255-20.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/14/2020] [Accepted: 03/25/2021] [Indexed: 11/21/2022] Open
Abstract
Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.
Collapse
Affiliation(s)
- Rhita Lamtahri
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| | - Mahmoud Hazime
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| | - Emma K Gowing
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, 76000, 9054, New Zealand
| | - Raghavendra Y Nagaraja
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, 76000, 9054, New Zealand
| | - Julie Maucotel
- Normandie Université, UNIROUEN, Animal Facility, Rouen, 76000, France
| | - Michael Alasoadura
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| | | | - Katia Lehongre
- Inserm U 1127, Centre National de la Recherche Scientifique Unite Mixte de Recherche 7225, Sorbonne Universités, UPMC Univ Paris 06 Unite Mixte de Recherche S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France
| | - Benjamin Lefranc
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Institute for Research and Innovation in Biomedicine, Normandie Université, PRIMACEN, Rouen, 76000, France
| | - Katarzyna Gach-Janczak
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Department of Biomolecular Chemistry, Medicinal University of Łódź, Łódź, 90-137, Poland
| | - Ann-Britt Marcher
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, 5230, Denmark
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, 5230, Denmark
| | - David Vaudry
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Institute for Research and Innovation in Biomedicine, Normandie Université, PRIMACEN, Rouen, 76000, France
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, 76000, 9054, New Zealand
| | - Jérôme Leprince
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
- Institute for Research and Innovation in Biomedicine, Normandie Université, PRIMACEN, Rouen, 76000, France
| | - Julien Chuquet
- Normandie Université, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1239, Neuronal and Neuroendocrine Differentiation and Communication, Rouen, France
| |
Collapse
|
16
|
Overview of the Neuroprotective Effects of the MAO-Inhibiting Antidepressant Phenelzine. Cell Mol Neurobiol 2021; 42:225-242. [PMID: 33839994 PMCID: PMC8732914 DOI: 10.1007/s10571-021-01078-3] [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: 12/29/2020] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
Phenelzine (PLZ) is a monoamine oxidase (MAO)-inhibiting antidepressant with anxiolytic properties. This multifaceted drug has a number of pharmacological and neurochemical effects in addition to inhibition of MAO, and findings on these effects have contributed to a body of evidence indicating that PLZ also has neuroprotective/neurorescue properties. These attributes are reviewed in this paper and include catabolism to the active metabolite β-phenylethylidenehydrazine (PEH) and effects of PLZ and PEH on the GABA-glutamate balance in brain, sequestration of reactive aldehydes, and inhibition of primary amine oxidase. Also discussed are the encouraging findings of the effects of PLZ in animal models of stroke, spinal cord injury, traumatic brain injury, and multiple sclerosis, as well other actions such as reduction of nitrative stress, reduction of the effects of a toxin on dopaminergic neurons, potential anticonvulsant actions, and effects on brain-derived neurotrophic factor, neural cell adhesion molecules, an anti-apoptotic factor, and brain levels of ornithine and N-acetylamino acids.
Collapse
|
17
|
Cerrah Gunes M, Gunes MS, Vural A, Aybuga F, Bayram A, Bayram KK, Sahin MI, Dogan ME, Ozdemir SY, Ozkul Y. Change in gene expression levels of GABA, glutamate and neurosteroid pathways due to acoustic trauma in the cochlea. J Neurogenet 2021; 35:45-57. [PMID: 33825593 DOI: 10.1080/01677063.2021.1904922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The characteristic feature of noise-induced hearing loss (NIHL) is the loss or malfunction of the outer hair cells (OHC) and the inner hair cells (IHC) of the cochlea. 90-95% of the spiral ganglion neurons, forming the cell bodies of cochlear nerve, synapse with the IHCs. Glutamate is the most potent excitatory neurotransmitter for IHC-auditory nerve synapses. Excessive release of glutamate in response to acoustic trauma (AT), may cause excitotoxicity by causing damage to the spiral ganglion neurons (SGN) or loss of the spiral ganglion dendrites, post-synaptic to the IHCs. Another neurotransmitter, GABA, plays an important role in the processing of acoustic stimuli and central regulation after peripheral injury, so it is potentially related to the regulation of hearing function and sensitivity after noise. The aim of this study is to evaluate the effect of AT on the expressions of glutamate excitotoxicity, GABA inhibition and neurosteroid synthesis genes.We exposed 24 BALB/c mice to AT. Controls were sacrificed without exposure to noise, Post-AT(1) and Post-AT(15) were sacrificed on the 1st and 15th day, respectively, after noise exposure. The expressions of various genes playing roles in glutamate, GABA and neurosteroid pathways were compared between groups by real-time PCR.Expressions of Cyp11a1, Gls, Gabra1, Grin2b, Sult1a1, Gad1, and Slc1a2 genes in Post-AT(15) mice were significantly decreased in comparison to control and Post-AT(1) mice. No significant differences in the expression of Slc6a1 and Slc17a8 genes was detected.These findings support the possible role of balance between glutamate excitotoxicity and GABA inhibition is disturbed during the post AT days and also the synthesis of some neurosteroids such as pregnenolone sulfate may be important in this balance.
Collapse
Affiliation(s)
- Meltem Cerrah Gunes
- Department of Medical Genetics, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Murat Salih Gunes
- Department of Otolaryngology, Izmit Seka State Hospital, Kocaeli, Turkey
| | - Alperen Vural
- Department of Otolaryngology, School of Medicine, Erciyes University, Kayseri, Turkey
| | | | - Arslan Bayram
- Etlik Zübeyde Hanım Women's Diseases Education and Research Hospital, Health Sciences University, T.R. Ministry of Health, Ankara, Turkey
| | - Keziban Korkmaz Bayram
- Department of Medical Genetics, School of Medicine, Yıldirim Beyazit University, Ankara, Turkey
| | - Mehmet Ilhan Sahin
- Department of Otolaryngology, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Muhammet Ensar Dogan
- Department of Medical Genetics, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Sevda Yesim Ozdemir
- Department of Medical Genetics, School of Medicine, Uskudar University, Istanbul, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, School of Medicine, Erciyes University, Kayseri, Turkey.,Center of Genome and Stem Cell, Kayseri, Turkey
| |
Collapse
|
18
|
Lin DJ, Cramer SC. Principles of Neural Repair and Their Application to Stroke Recovery Trials. Semin Neurol 2021; 41:157-166. [PMID: 33663003 DOI: 10.1055/s-0041-1725140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Neural repair is the underlying therapeutic strategy for many treatments currently under investigation to improve recovery after stroke. Repair-based therapies are distinct from acute stroke strategies: instead of salvaging threatened brain tissue, the goal is to improve behavioral outcomes on the basis of experience-dependent brain plasticity. Furthermore, timing, concomitant behavioral experiences, modality specific outcome measures, and careful patient selection are fundamental concepts for stroke recovery trials that can be deduced from principles of neural repair. Here we discuss core principles of neural repair and their implications for stroke recovery trials, highlighting related issues from key studies in humans. Research suggests a future in which neural repair therapies are personalized based on measures of brain structure and function, genetics, and lifestyle factors.
Collapse
Affiliation(s)
- David J Lin
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts.,VA RR&D Center for Neurorestoration and Neurotechnology, Rehabilitation R&D Service, Department of VA Medical Center, Providence, Rhode Island
| | - Steven C Cramer
- Department of Neurology, University of California, Los Angeles, California.,California Rehabilitation Institute, Los Angeles, California
| |
Collapse
|
19
|
Xiao G, Lyu M, Li Z, Cao L, Liu X, Wang Y, He S, Chen Z, Du H, Feng Y, Wang J, Zhu Y. Restoration of early deficiency of axonal guidance signaling by guanxinning injection as a novel therapeutic option for acute ischemic stroke. Pharmacol Res 2021; 165:105460. [PMID: 33513357 DOI: 10.1016/j.phrs.2021.105460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/22/2021] [Indexed: 01/12/2023]
Abstract
Despite of its high morbidity and mortality, there is still a lack of effective treatment for ischemic stroke in part due to our incomplete understanding of molecular mechanisms of its pathogenesis. In this study, we demonstrate that SHH-PTCH1-GLI1-mediated axonal guidance signaling and its related neurogenesis, a central pathway for neuronal development, also plays a critical role in early stage of an acute stroke model. Specifically, in vivo, we evaluated the effect of GXNI on ischemic stroke mice via using the middle cerebral artery embolization model, and found that GXNI significantly alleviated cerebral ischemic reperfusion (I/R) injury by reducing the volume of cerebral infarction, neurological deficit score and cerebral edema, reversing the BBB permeability and histopathological changes. A combined approach of RNA-seq and network pharmacology analysis was used to reveal the underlying mechanisms of GXNI followed by RT-PCR, immunohistochemistry and western blotting validation. It was pointed out that axon guidance signaling pathway played the most prominent role in GXNI action with Shh, Ptch1, and Gli1 genes as the critical contributors in brain protection. In addition, GXNI markedly prevented primary cortical neuron cells from oxygen-glucose deprivation/reoxygenation damage in vitro, and promoted axon growth and synaptogenesis of damaged neurons, which further confirmed the results of in vivo experiments. Moreover, due to the inhibition of the SHH-PTCH1-GLI1 signaling pathway by cyclopropylamine, the effect of GXNI was significantly weakened. Hence, our study provides a novel option for the clinical treatment of acute ischemic stroke by GXNI via SHH-PTCH1-GLI1-mediated axonal guidance signaling, a neuronal development pathway previously considered for after-stroke recovery.
Collapse
Affiliation(s)
- Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Ming Lyu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhixiong Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Linghua Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Zihao Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Hongxia Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Jigang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China.
| |
Collapse
|
20
|
Sitruk-Ware R, Bonsack B, Brinton R, Schumacher M, Kumar N, Lee JY, Castelli V, Corey S, Coats A, Sadanandan N, Gonzales-Portillo B, Heyck M, Shear A, Blaise C, Zhang H, Sheyner M, García-Sánchez J, Navarro L, El-Etr M, De Nicola AF, Borlongan CV. Progress in progestin-based therapies for neurological disorders. Neurosci Biobehav Rev 2020; 122:38-65. [PMID: 33359391 DOI: 10.1016/j.neubiorev.2020.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/26/2020] [Accepted: 12/12/2020] [Indexed: 12/16/2022]
Abstract
Hormone therapy, primarily progesterone and progestins, for central nervous system (CNS) disorders represents an emerging field of regenerative medicine. Following a failed clinical trial of progesterone for traumatic brain injury treatment, attention has shifted to the progestin Nestorone for its ability to potently and selectively transactivate progesterone receptors at relatively low doses, resulting in robust neurogenetic, remyelinating, and anti-inflammatory effects. That CNS disorders, including multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), spinal cord injury (SCI), and stroke, develop via demyelinating, cell death, and/or inflammatory pathological pathways advances Nestorone as an auspicious candidate for these disorders. Here, we assess the scientific and clinical progress over decades of research into progesterone, progestins, and Nestorone as neuroprotective agents in MS, ALS, SCI, and stroke. We also offer recommendations for optimizing timing, dosage, and route of the drug regimen, and identifying candidate patient populations, in advancing Nestorone to the clinic.
Collapse
Affiliation(s)
| | - Brooke Bonsack
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Vanessa Castelli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Alexandreya Coats
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Nadia Sadanandan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bella Gonzales-Portillo
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Matt Heyck
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Alex Shear
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cozene Blaise
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Henry Zhang
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michael Sheyner
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Julián García-Sánchez
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Lisset Navarro
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
21
|
Zong X, Li Y, Liu C, Qi W, Han D, Tucker L, Dong Y, Hu S, Yan X, Zhang Q. Theta-burst transcranial magnetic stimulation promotes stroke recovery by vascular protection and neovascularization. Theranostics 2020; 10:12090-12110. [PMID: 33204331 PMCID: PMC7667689 DOI: 10.7150/thno.51573] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/26/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale: The integrity and function of the blood-brain barrier (BBB) is compromised after stroke. The current study was performed to examine potential beneficial effects and underlying mechanisms of repetitive transcranial magnetic stimulation (rTMS) on angiogenesis and vascular protection, function, and repair following stroke, which are largely unknown. Methods: Using a rat photothrombotic (PT) stroke model, continuous theta-burst rTMS was administered once daily to the infarcted hemisphere for 5 min, beginning 3 h after PT stroke. This treatment was applied for 6 days. BBB integrity, blood flow, vascular associated proteins, angiogenesis, integrity of neuronal morphology and structure, and behavioral outcome were measured and analyzed at 6 and/or 22 days after PT stroke. Results: We report that rTMS significantly mitigated BBB permeabilization and preserved important BBB components ZO-1, claudin-5, occludin, and caveolin-1 from PT-induced degradation. Damage to vascular structure, morphology, and perfusion was ameliorated by rTMS, resulting in improved local tissue oxygenation. This was accompanied with robust protection of critical vascular components and upregulation of regulatory factors. A complex cytokine response was induced by PT, particularly at the late phase. Application of rTMS modulated this response, ameliorating levels of cytokines related to peripheral immune cell infiltration. Further investigation revealed that rTMS promoted and sustained post-ischemic angiogenesis long-term and reduced apoptosis of newborn and existing vascular endothelial cells. Application of rTMS also inhibited PT-induced excessive astrocyte-vasculature interactions and stimulated an A1 to A2 shift in vessel-associated astrocytes. Mechanistic studies revealed that rTMS dramatically increased levels of PDGFRβ associated with A2 astrocytes and their adjacent vasculature. As well, A2 astrocytes displayed marked amplification of the angiogenesis-related factors VEGF and TGFβ. PT induced a rise in vessel-associated expression of HIF-1α that was starkly intensified by rTMS treatment. Finally, rTMS preserved neuronal morphology, synaptic structure integrity and behavioral outcome. Conclusions: These results indicate that rTMS can exert powerful protective and restorative effects on the peri-infarct microvasculature after PT stroke by, in part, promoting HIF-1α signaling and shifting vessel-associated astrocytic polarization to the A2 phenotype. This study provides further support for the potent protective effects of rTMS in the context of ischemic stroke, and these findings implicate vascular repair and protection as an important underlying phenomenon.
Collapse
|
22
|
Tang Z, Yang F, Dong Y, Ma C, Sun S, Shan Y, Zhang Y, Liu H. Midazolam contributes to neuroprotection against hypoxia/reoxygenation-induced brain injury in neonatal rats via regulation of EAAT2. Brain Res Bull 2020; 161:136-146. [DOI: 10.1016/j.brainresbull.2020.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/18/2020] [Accepted: 04/20/2020] [Indexed: 01/22/2023]
|
23
|
Herzog C, Greenald D, Larraz J, Keatinge M, Herrgen L. RNA-seq analysis and compound screening highlight multiple signalling pathways regulating secondary cell death after acute CNS injury in vivo. Biol Open 2020; 9:9/5/bio050260. [PMID: 32366533 PMCID: PMC7225090 DOI: 10.1242/bio.050260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Understanding the molecular mechanisms that regulate secondary cell death after acute central nervous system (CNS) injury is critical for the development of effective neuroprotective drugs. Previous research has shown that neurotoxic processes including excitotoxicity, oxidative stress and neuroinflammation can cause secondary cell death. Nevertheless, clinical trials targeting these processes have been largely unsuccessful, suggesting that the signalling pathways underlying secondary cell death remain incompletely understood. Due to their suitability for live imaging and their amenability to genetic and pharmacological manipulation, larval zebrafish provide an ideal platform for studying the regulation of secondary cell death in vivo Here, we use RNA-seq gene expression profiling and compound screening to identify signalling pathways that regulate secondary cell death after acute neural injury in larval zebrafish. RNA-seq analysis of genes upregulated in cephalic mpeg1+ macrophage-lineage cells isolated from mpeg1:GFP transgenic larvae after neural injury suggested an involvement of cytokine and polyamine signalling in secondary cell death. Furthermore, screening a library of FDA approved compounds indicated roles for GABA, serotonin and dopamine signalling. Overall, our results highlight multiple signalling pathways that regulate secondary cell death in vivo, and thus provide a starting point for the development of novel neuroprotective treatments for patients with CNS injury.This article has an associated First Person interview with the two first authors of the paper.
Collapse
Affiliation(s)
- Chiara Herzog
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - David Greenald
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Juan Larraz
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Leah Herrgen
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
24
|
Łątka K, Jończyk J, Bajda M. γ-Aminobutyric acid transporters as relevant biological target: Their function, structure, inhibitors and role in the therapy of different diseases. Int J Biol Macromol 2020; 158:S0141-8130(20)32987-1. [PMID: 32360967 DOI: 10.1016/j.ijbiomac.2020.04.126] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/13/2022]
Abstract
γ-Aminobutyric acid (GABA) is a major inhibitory neurotransmitter in the nervous system. It plays a crucial role in many physiological processes. Upon release from the presynaptic element, it is removed from the synaptic cleft by reuptake due to the action of GABA transporters (GATs). GATs belong to a large SLC6 protein family whose characteristic feature is sodium-dependent relocation of neurotransmitters through the cell membrane. GABA transporters are characterized in many contexts, but their spatial structure is not fully known. They are divided into four types, which differ in occurrence and role. Herein, the special attention was paid to these transporting proteins. This comprehensive review presents the current knowledge about GABA transporters. Their distribution in the body, physiological functions and possible utilization in the therapy of different diseases were fully discussed. The important structural features were described based on published data, including sequence analysis, mutagenesis studies, and comparison with known SLC6 transporters for leucine (LeuT), dopamine (DAT) and serotonin (SERT). Moreover, the most important inhibitors of GABA transporters of various basic scaffolds, diverse selectivity and potency were presented.
Collapse
Affiliation(s)
- Kamil Łątka
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Physicochemical Drug Analysis, 30-688 Cracow, Medyczna 9, Poland
| | - Jakub Jończyk
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Physicochemical Drug Analysis, 30-688 Cracow, Medyczna 9, Poland
| | - Marek Bajda
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Physicochemical Drug Analysis, 30-688 Cracow, Medyczna 9, Poland.
| |
Collapse
|
25
|
Park KT, Han JK, Kim SJ, Lim YH. Gamma-Aminobutyric Acid Increases Erythropoietin by Activation of Citrate Cycle and Stimulation of Hypoxia-Inducible Factors Expression in Rats. Biomolecules 2020; 10:E595. [PMID: 32290638 PMCID: PMC7226430 DOI: 10.3390/biom10040595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/04/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (EPO) is the primary regulator of erythropoiesis in the mammalian fetus and adult. Deficiency of EPO induces anemia. In this study, we investigated the effect of gamma-aminobutyric acid (GABA) on serum EPO levels and erythropoiesis in rats. Expression levels of Epo-related genes were measured by quantitative real-time PCR (qPCR) and expression of Epo and Epo receptor (Epor) proteins were measured by immunohistochemistry. The gene and protein expression profiles of kidney tissue in GABA-treated rats were evaluated by ribonucleic acid (RNA) sequencing and two-dimensional electrophoresis (2-DE), respectively. GABA significantly increased serum EPO levels and expression levels of Epo and Epor. GABA increased expression levels of hypoxia-inducible factor (Hif)-1 and Hif-2. Seven proteins with expression levels showing >2-fold change were identified by 2-DE followed by MALDI-TOF MS in GABA-treated rat kidney. The top KEGG pathway from the identified proteins was the tricarboxylic acid cycle, and nicotinamide adenine dinucleotide (NADH) dehydrogenase, succinate dehydrogenase, and isocitrate dehydrogenase were identified as key proteins. GABA treatment significantly increased ATP levels and NADH dehydrogenase activity in a dose-dependent manner. In conclusion, GABA shows a new physiological role in EPO production, and it can thus can contribute to the prevention of anemia when used alone or in combination with other anemia treating drugs.
Collapse
Affiliation(s)
- Keun-Tae Park
- Research and Development Center, Milae Bioresources Co., Ltd., Seoul 05836, Korea; (K.-T.P.); (J.-K.H.); (S.J.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Korea
| | - Jong-Kwon Han
- Research and Development Center, Milae Bioresources Co., Ltd., Seoul 05836, Korea; (K.-T.P.); (J.-K.H.); (S.J.K.)
| | - Seong Jin Kim
- Research and Development Center, Milae Bioresources Co., Ltd., Seoul 05836, Korea; (K.-T.P.); (J.-K.H.); (S.J.K.)
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Korea
- Department of Public Health Science (BK21 PLUS Program), Graduate School, Korea University, Seoul 02841, Korea
- Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 08308, Korea
| |
Collapse
|
26
|
Optogenetic translocation of protons out of penumbral neurons is protective in a rodent model of focal cerebral ischemia. Brain Stimul 2020; 13:881-890. [PMID: 32289721 DOI: 10.1016/j.brs.2020.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Intracellular acidosis in the ischemic penumbra can contribute to further cell death, effectively enlarging the infarct core. Restoring the acid-base balance may enhance tissue survivability after cerebral ischemia. OBJECTIVE This study investigated whether translocating protons out of penumbral neurons could mitigate tissue acidification and induce neuroprotection in a rodent model of acute cerebral ischemia. METHODS We modulated the penumbral neurons via a light-driven pump to translocate protons out (i.e., archaerhodopsin/ArchT group) or into (i.e., channelrhodopsin-2/ChR2 group) neurons after focal cerebral ischemia in rats. Intracellular pH values were imaged via neutral red (NR) fluorescence and cerebral blood flow (CBF) was monitored through laser speckle contrast imaging (LSCI). Global CBF responses to electrical stimulation of the hindlimbs were obtained 24 h and 48 h after ischemia to assess neurological function. Behavioral and histological outcomes were evaluated 48 h after ischemia. A control group without gene modification was included. RESULTS The reduction of relative pH (RpH), the amplitude of negative peak of hypoemic response (RNP) and the hemispheric lateralization index (LI) in ArchT group were significantly less than those of the ChR2 or control group. Moreover, RpH was strongly correlated with RNP (r = 0.60) and LI (r24h = 0.80, r48h = 0.59). In addition, behavioral and histological results supported a neuroprotective effect of countering neuronal acidosis in penumbra through optogenetic stimulation. CONCLUSION(S) These results indicate that countering intracellular acidosis by optogenetically translocating protons out of penumbral neurons during the acute ischemic stage could induce protection after ischemic brain injury.
Collapse
|
27
|
Feng YW, Huang YQ, Yan Y, Li G, He XF, Liang FY, Pei Z, Lan Y, Xu GQ. Phasic GABA signaling mediates the protective effects of cTBS against cerebral ischemia in mice. Neurosci Lett 2019; 715:134611. [PMID: 31698026 DOI: 10.1016/j.neulet.2019.134611] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 09/25/2019] [Accepted: 11/03/2019] [Indexed: 01/26/2023]
Abstract
Continuous theta burst stimulation (cTBS) has been widely recognized as a therapeutic treatment for ischemic stroke, but the underlying mechanism is still elusive. Here, we investigated the protective effects of cTBS in the posterior parietal cortex during the chronic phase of stroke in the photothrombotic ischemic model. Infarction volume and neuron excitability in the peri-infarct area were assessed using immunohistochemistry and whole-cell patch-clamp. Spatial cognitive function was measured using the Morris water maze. Gamma-Amino butyric acid (GABA) interneurons were responsive to cTBS, and cTBS induced elevated phasic inhibition rather than tonic inhibition. Given that GABA-A-mediated phasic inhibition was elevated during the chronic phase of ischemic stroke for 30 days and was beneficial for stroke recovery, we investigated the therapeutic potential of cTBS in promoting functional recovery and found that the elevated phasic inhibition by cTBS improved spatial cognitive function in the photothrombotic stroke mouse model with induction in the posterior parietal cortex. Our study indicates the mechanism by which cTBS may modify the excitability of the brain cortex and provides novel insight into the potential of cTBS to protect against neuronal dysfunction in ischemic stroke.
Collapse
Affiliation(s)
- Yi-Wei Feng
- Department of Rehabilitation Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China; Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi-Qing Huang
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Rehabilitation Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Yan
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Xiao-Fei He
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Feng-Yin Liang
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Pei
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yue Lan
- Department of Rehabilitation Medicine, Guangzhou First People's Hospital, Guangzhou, China; Department of Rehabilitation Medicine, The Second Affiliated Hospital of South China University of Technology, Guangzhou, China.
| | - Guang-Qing Xu
- Department of Rehabilitation Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
28
|
Wang H, Cheng X, Yu H, Zhang X, Guan M, Zhao L, Liu Y, Linag Y, Luo Y, Zhao C. Activation of GABAA receptors enhances the behavioral recovery but not axonal sprouting in ischemic rats. Restor Neurol Neurosci 2019; 37:315-331. [PMID: 31227671 DOI: 10.3233/rnn-180827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Huibin Wang
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xi Cheng
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hang Yu
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Xiuchun Zhang
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Meiting Guan
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Lanqing Zhao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yang Liu
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yifan Linag
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yujia Luo
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Chuansheng Zhao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
29
|
Cragg JJ, Tong B, Jutzeler CR, Warner FM, Cashman N, Geisler F, Kramer JLK. A Longitudinal Study of the Neurologic Safety of Acute Baclofen Use After Spinal Cord Injury. Neurotherapeutics 2019; 16:858-867. [PMID: 30725362 PMCID: PMC6694358 DOI: 10.1007/s13311-019-00713-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The objective of our study was to determine whether treatment with baclofen is neurologically safe with respect to exposure during recovery from spinal cord injury. We performed a secondary longitudinal analysis of a cohort of adult patients with traumatic acute spinal cord injury. Cumulative baclofen dose was computed over the first 4 weeks following injury from concomitant medication information from a completed clinical trial. The main outcome measure was neurologic status, which was assessed over 52 weeks with "marked recovery" defined as the conversion to higher sensory and motor function. To complete the drug safety profile, drug toxicity was assessed with assays from standard blood work. Multivariable Cox regression was used to compute hazard ratios (HRs) and 95% confidence intervals (CIs). Of the cohort (n = 651), 18% (n = 115) received baclofen within 4 weeks post injury. Baclofen use was associated with higher rates of marked neurologic recovery, even after adjustment for injury severity (HR = 2.1, 95% CI 1.5-3.0 for high dose vs none). Baclofen exposure was not associated with liver or renal side effects. The use of other medications indicated for spasticity was not associated with neurological outcomes. Overall, this longitudinal analysis provides level 3 evidence on the neurologic safety of baclofen and potential beneficial effects on recovery in the early days after acute traumatic spinal cord injury. The usefulness of concomitant medication files from completed clinical trials is highlighted. We also highlight the importance of incorporating logical patient questions and neurological outcomes into research addressing drug safety.
Collapse
Affiliation(s)
- Jacquelyn J Cragg
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, 818 West 10th Avenue, Vancouver, British Columbia, V5Z 1M9, Canada.
| | - Bobo Tong
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, 818 West 10th Avenue, Vancouver, British Columbia, V5Z 1M9, Canada
| | - Catherine R Jutzeler
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, 818 West 10th Avenue, Vancouver, British Columbia, V5Z 1M9, Canada
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Freda M Warner
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, 818 West 10th Avenue, Vancouver, British Columbia, V5Z 1M9, Canada
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neil Cashman
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fred Geisler
- University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John L K Kramer
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, 818 West 10th Avenue, Vancouver, British Columbia, V5Z 1M9, Canada
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Kim GA, Gan’shina TS, Kurza EV, Kurdyumov IN, Maslennikov DV, Mirzoian RS. New cerebrovascular agent with hypotensive activity. RESEARCH RESULTS IN PHARMACOLOGY 2019. [DOI: 10.3897/rrpharmacology.5.35392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: In cerebrovascular disorders, special attention is paid to a hypertensive cerebrovascular crisis, which combines a vascular injury of the brain and hypertension. The paper studies the cerebrovascular properties of the calcium channel blocker of S-Amlodipine nicotinate antihypertensive agent.
Materials and methods: Tests were performed on 96 nonlinear male rats, measuring local blood flow in the cerebral cortex in 36 awake animals, using a laser Doppler flowmeter. Cerebral circulation was recorded in the animals when modeling ischemic and hemorrhagic brain injuries.
Results and discussion: S-Amlodipine nicotinate (0.1 mg/kg i/v) shows a pronounced cerebrovascular activity in the models of ischemic and hemorrhagic injuries of the brain. In terms of the vasodilating effect in ischemic brain injury, the drug is comparable to mexidol, nimodipine, picamilon, but is superior to nimodipine and picamilon in terms of duration of action, and in the model of hemorrhagic stroke, S-Amlodipine nicotinate is superior to nimodipine and is comparable to picamilon and mexidol. The analysis of the mechanism of action of the agent revealed the participation of GABA A-receptors in the implementation of cerebrovascular properties of the agent.
Conclusion: Significant cerebrovascular activity of S-Amlodipine nicotinate (0.1 mg/kg i/v) antihypertensive agent was revealed. The presence of GABAergic mechanism on cerebral blood flow in the agent action along with blockade of slow calcium channels ensures its high efficacy in treatment of both ischemic and hemorrhagic brain injuries.
Collapse
|
31
|
|
32
|
Baker G, Matveychuk D, MacKenzie EM, Holt A, Wang Y, Kar S. Attenuation of the effects of oxidative stress by the MAO-inhibiting antidepressant and carbonyl scavenger phenelzine. Chem Biol Interact 2019; 304:139-147. [PMID: 30857888 DOI: 10.1016/j.cbi.2019.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/21/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
Abstract
Phenelzine (β-phenylethylhydrazine) is a monoamine oxidase (MAO)-inhibiting antidepressant with anxiolytic properties. It possesses a number of important pharmacological properties which may alter the effects of oxidative stress. After conducting a comprehensive literature search, the authors of this review paper aim to provide an overview and discussion of the mechanisms by which phenelzine may attenuate oxidative stress. It inhibits γ-aminobutyric acid (GABA) transaminase, resulting in elevated brain GABA levels, inhibits both MAO and primary amine oxidase and, due to its hydrazine-containing structure, reacts chemically to sequester a number of reactive aldehydes (e.g. acrolein and 4-hydroxy-2-nonenal) proposed to be implicated in oxidative stress in a number of neurodegenerative disorders. Phenelzine is unusual in that it is both an inhibitor of and a substrate for MAO, the latter action producing at least one active metabolite, β-phenylethylidenehydrazine (PEH). This metabolite inhibits GABA transaminase, is a very weak inhibitor of MAO but a strong inhibitor of primary amine oxidase, and sequesters aldehydes. Phenelzine may ameliorate the effects of oxidative stress by reducing formation of reactive metabolites (aldehydes, hydrogen peroxide, ammonia/ammonia derivatives) produced by the interaction of MAO with biogenic amines, by sequestering various other reactive aldehydes and by inhibiting primary amine oxidase. In PC12 cells treated with the neurotoxin MPP+, phenelzine has been reported to reduce several adverse effects of MPP+. It has also been reported to reduce lipid peroxidative damage induced in plasma and platelet proteins by peroxynitrite. In animal models, phenelzine has a neuroprotective effect in global ischemia and in cortical impact traumatic brain injury. Recent studies reported in the literature on the possible involvement of acrolein in spinal cord injury and multiple sclerosis indicate that phenelzine can attenuate adverse effects of acrolein in these models. Results from studies in our laboratories on effects of phenelzine and PEH on primary amine oxidase (which catalyzes formation of toxic aldehydes and is overexpressed in Alzheimer's disease), on sequestration of the toxic aldehyde acrolein, and on reduction of acrolein-induced toxicity in mouse cortical neurons are also reported.
Collapse
Affiliation(s)
- Glen Baker
- Department of Psychiatry (Neurochemical Research Unit), University of Alberta, Edmonton, Canada.
| | - Dmitriy Matveychuk
- Department of Psychiatry (Neurochemical Research Unit), University of Alberta, Edmonton, Canada.
| | - Erin M MacKenzie
- Department of Psychiatry (Neurochemical Research Unit), University of Alberta, Edmonton, Canada.
| | - Andrew Holt
- Department of Psychiatry (Neurochemical Research Unit), University of Alberta, Edmonton, Canada.
| | - Yanlin Wang
- Department of Psychiatry (Neurochemical Research Unit), University of Alberta, Edmonton, Canada; Department of Medicine (Neurology), University of Alberta, Edmonton, Canada.
| | - Satyabrata Kar
- Department of Psychiatry (Neurochemical Research Unit), University of Alberta, Edmonton, Canada; Department of Medicine (Neurology), University of Alberta, Edmonton, Canada.
| |
Collapse
|
33
|
Delayed treatment of α5 GABAA receptor inverse agonist improves functional recovery by enhancing neurogenesis after cerebral ischemia-reperfusion injury in rat MCAO model. Sci Rep 2019; 9:2287. [PMID: 30783142 PMCID: PMC6381084 DOI: 10.1038/s41598-019-38750-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/18/2018] [Indexed: 11/08/2022] Open
Abstract
Development of effective therapeutics and treatment strategy to promote recovery after cerebral ischemia-reperfusion injury necessitates further understandings of the complex pathophysiology of ischemic stroke. Given that α5-GABAAR inhibition has been shown to be involved in functional recovery after stroke, the present study was designed to evaluate the effects of treatment timing of α5 GABAAR inhibition on post-middle cerebral artery occlusion (MCAO) functional recovery. To this end, we examined the effects of L655,708 (α5 GABAAR inverse agonist) treatment at 3 or 7 days post-ischemia on apoptosis and neurogenesis in the peri-infarct region, brain infarction size, as well as modified neurological severity score (mNSS) and rotarod test time in rats. Consistent with previous reports, we found that when the treatment of L655,708 was initiated at post-MCAO day 3, it did not alter the functional recovery in rats. However, when the treatment of L655,708 was initiated at post-MCAO day 7, it demonstrated beneficial effects on functional recovery in rats. Interestingly, this phenomenon was not associated with altered brain infarction size nor with changes in brain cell apoptosis. However, we found that delayed treatment of L655,708 at post-MCAO day 7 significantly increased neurogenesis in peri-infarct zone in rats. These results suggested that removing α5 GABAAR-mediated tonic inhibition after cerebral ischemia-reperfusion injury may be an effective therapeutic strategy for promoting functional recovery from stroke.
Collapse
|
34
|
Hollas MA, Ben Aissa M, Lee SH, Gordon-Blake JM, Thatcher GRJ. Pharmacological manipulation of cGMP and NO/cGMP in CNS drug discovery. Nitric Oxide 2019; 82:59-74. [PMID: 30394348 PMCID: PMC7645969 DOI: 10.1016/j.niox.2018.10.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/14/2018] [Accepted: 10/25/2018] [Indexed: 12/21/2022]
Abstract
The development of small molecule modulators of NO/cGMP signaling for use in the CNS has lagged far behind the use of such clinical agents in the periphery, despite the central role played by NO/cGMP in learning and memory, and the substantial evidence that this signaling pathway is perturbed in neurodegenerative disorders, including Alzheimer's disease. The NO-chimeras, NMZ and Nitrosynapsin, have yielded beneficial and disease-modifying responses in multiple preclinical animal models, acting on GABAA and NMDA receptors, respectively, providing additional mechanisms of action relevant to synaptic and neuronal dysfunction. Several inhibitors of cGMP-specific phosphodiesterases (PDE) have replicated some of the actions of these NO-chimeras in the CNS. There is no evidence that nitrate tolerance is a phenomenon relevant to the CNS actions of NO-chimeras, and studies on nitroglycerin in the periphery continue to challenge the dogma of nitrate tolerance mechanisms. Hybrid nitrates have shown much promise in the periphery and CNS, but to date only one treatment has received FDA approval, for glaucoma. The potential for allosteric modulation of soluble guanylate cyclase (sGC) in brain disorders has not yet been fully explored nor exploited; whereas multiple applications of PDE inhibitors have been explored and many have stalled in clinical trials.
Collapse
Affiliation(s)
- Michael A Hollas
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Manel Ben Aissa
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Sue H Lee
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Jesse M Gordon-Blake
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA.
| |
Collapse
|
35
|
El Amki M, Binder N, Steffen R, Schneider H, Luft AR, Weller M, Imthurn B, Merki-Feld GS, Wegener S. Contraceptive drugs mitigate experimental stroke-induced brain injury. Cardiovasc Res 2018; 115:637-646. [DOI: 10.1093/cvr/cvy248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
AbstractAimsEffective stroke treatments beyond reperfusion remain scant. The natural steroid hormone progesterone has shown protective effects in experimental models of brain injury and cardiovascular disease. However, unfavourable bioavailability limits its clinical use. Desogestrel and drospirenone are new generation progestins with progesterone-like properties, developed as oral contraceptives with excellent bioavailability and safety profile. We investigated the neuroprotective properties of these progestins in vivo using transient middle cerebral artery occlusion (MCAO) and in vitro using an oxygen-glucose deprivation and reoxygenation (OGD/R) model in primary neuronal cells.Methods and resultsMCAO was induced in female, female ovariectomized (modelling postmenopausal females) and male mice. Treatment with the progestins resulted in less severe strokes after MCAO and less neuronal death in OGD/R. Desogestrel and drospirenone induced higher expression levels of GABAAR α4 and delta subunits within the brain, suggesting changes in GABAAR configuration favouring tonic inhibition as potential mechanism of action. Treatment with the GABAAR blocker picrotoxin abolished the protection afforded by the progestins in vivo and in vitro.ConclusionFor the first time, here, we delineate a potential role of desogestrel and drospirenone, both clinically approved and safe drugs in mitigating the consequences of stroke. Contraception with desogestrel and drospirenone in progestin-only preparations may be particularly beneficial for women at risk of stroke.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Nadine Binder
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Riccardo Steffen
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Hannah Schneider
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Andreas R Luft
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Bruno Imthurn
- Department of Reproductive Endocrinology, University Hospital Zurich, Switzerland
| | | | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| |
Collapse
|
36
|
Durdag E, Yildirim Z, Unlu NL, Kale A, Ceviker N. Neuroprotective Effects of Vigabatrin on Spinal Cord Ischemia-Reperfusion Injury. World Neurosurg 2018; 120:e33-e41. [PMID: 30031958 DOI: 10.1016/j.wneu.2018.07.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Spinal cord ischemia is a serious and catastrophic clinicopathologic condition. Despite studies reported over the last 20 years, alternative and efficient treatment options remain unclear. We examined the neuroprotective effects of vigabatrin on a spinal ischemia-reperfusion model. METHODS We divided 24 New Zealand rabbits into 4 groups (control, ischemia reperfusion, and low-dose and high-dose vigabatrin). The control group underwent only abdominal surgery, whereas an abdominal aortic cross-clamp model of spinal ischemia was performed in the other groups. Clips were removed after 30 minutes and 50 and 150 mg/kg vigabatrin was administered intraperitoneally to the low-dose and high-dose groups, respectively. Neurologic examination was performed for 48 hours, after which the rabbits were sacrificed and a blood sample obtained. Biochemical examination of malondialdehyde, advanced oxidation protein products, total nitric oxide, and glutathione levels and superoxide dismutase activities in plasma and tissue sample, and histopathologic examination of the spinal cord were performed and statistical results compared between the groups. RESULTS Low-dose vigabatrin had statistically significant effects of neuroprotection on spinal ischemia. Although high-dose vigabatrin had similar effects, the results were not statistically significant for all parameters of biochemical analysis. In addition, histopathologic examination showed some toxic effects of high-dose vigabatrin. CONCLUSIONS Neuroprotective effects of vigabatrin are shown. For clinical use, further studies are needed.
Collapse
Affiliation(s)
- Emre Durdag
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - Zuhal Yildirim
- Etimesgut Public Health Laboratory, Etimesgut, Ankara, Turkey.
| | - Nese Lortlar Unlu
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Aydemir Kale
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - Necdet Ceviker
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| |
Collapse
|
37
|
Landucci E, Llorente IL, Anuncibay-Soto B, Pellegrini-Giampietro DE, Fernández-López A. Bicuculline Reverts the Neuroprotective Effects of Meloxicam in an Oxygen and Glucose Deprivation (OGD) Model of Organotypic Hippocampal Slice Cultures. Neuroscience 2018; 386:68-78. [PMID: 29949743 DOI: 10.1016/j.neuroscience.2018.06.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/25/2018] [Accepted: 06/18/2018] [Indexed: 01/17/2023]
Abstract
We previously demonstrated that the non-steroidal anti-inflammatory agent meloxicam has neuroprotective effects in an oxygen and glucose deprivation model (OGD) of rat organotypic hippocampal slice cultures. We wondered if GABAergic transmission changed the neuroprotective effects of meloxicam and if meloxicam was able to modulate endoplasmic reticulum stress (ER stress) in this model. Mortality was measured using propidium iodide. Western blot assays were performed to measure levels of cleaved and non-cleaved caspase-3 to quantify apoptosis, while levels of GRP78, GRP94 and phosphorylated eIF2α were used to detect unfolded protein response (UPR). Transcript levels of GRP78, GRP94 and GABAergic receptor α, β, and γ subunits were measured by real-time quantitative polymerase chain reaction (qPCR). In the present study, we show that the presence of meloxicam in a 30 min OGD assay, followed by 24 h of normoxic conditions, presented an antiapoptotic effect. The simultaneous presence of the GABAA receptor antagonist, bicuculline, in combination with meloxicam blocked the neuroprotective effect provided by the latter. However, in light of its effects on caspase 3 and PARP, bicuculline did not seem to promote the apoptotic pathway. Our results also showed that meloxicam modified the unfolded protein response (UPR), as well as the transcriptional response of different genes, including the GABAA receptor, alpha1, beta3 and gamma2 subunits. We concluded that meloxicam has a neuroprotective anti-apoptotic action, is able to enhance the UPR independently of the systemic anti-inflammatory response and its neuroprotective effect can be inhibited by blocking GABAA receptors.
Collapse
Affiliation(s)
- Elisa Landucci
- Sezione di Farmacologia Clinica e Oncologia, Dipartimento di Scienze della Salute, Università di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Irene L Llorente
- Neurology Department, David Geffen School of Medicine, University of California, Los Angeles, USA.
| | - Berta Anuncibay-Soto
- Área de Biología Celular, Instituto de Biomedicina, Universidad de León, 24071 León, Spain; Neural Therapies SL, Edificio Institutos de Investigación, Local B14, Universidad de León, 24071 León, Spain.
| | - Domenico E Pellegrini-Giampietro
- Sezione di Farmacologia Clinica e Oncologia, Dipartimento di Scienze della Salute, Università di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy.
| | | |
Collapse
|
38
|
García-Berrocoso T, Llombart V, Colàs-Campàs L, Hainard A, Licker V, Penalba A, Ramiro L, Simats A, Bustamante A, Martínez-Saez E, Canals F, Sanchez JC, Montaner J. Single Cell Immuno-Laser Microdissection Coupled to Label-Free Proteomics to Reveal the Proteotypes of Human Brain Cells After Ischemia. Mol Cell Proteomics 2017; 17:175-189. [PMID: 29133510 DOI: 10.1074/mcp.ra117.000419] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Indexed: 12/13/2022] Open
Abstract
Cerebral ischemia entails rapid tissue damage in the affected brain area causing devastating neurological dysfunction. How each component of the neurovascular unit contributes or responds to the ischemic insult in the context of the human brain has not been solved yet. Thus, the analysis of the proteome is a straightforward approach to unraveling these cell proteotypes. In this study, post-mortem brain slices from ischemic stroke patients were obtained corresponding to infarcted (IC) and contralateral (CL) areas. By means of laser microdissection, neurons and blood brain barrier structures (BBB) were isolated and analyzed using label-free quantification. MS data are available via ProteomeXchange with identifier PXD003519. Ninety proteins were identified only in neurons, 260 proteins only in the BBB and 261 proteins in both cell types. Bioinformatics analyses revealed that repair processes, mainly related to synaptic plasticity, are outlined in microdissected neurons, with nonexclusive important functions found in the BBB. A total of 30 proteins showing p < 0.05 and fold-change> 2 between IC and CL areas were considered meaningful in this study: 13 in neurons, 14 in the BBB and 3 in both cell types. Twelve of these proteins were selected as candidates and analyzed by immunohistofluorescence in independent brains. The MS findings were completely verified for neuronal SAHH2 and SRSF1 whereas the presence in both cell types of GABT and EAA2 was only validated in neurons. In addition, SAHH2 showed its potential as a prognostic biomarker of neurological improvement when analyzed early in the plasma of ischemic stroke patients. Therefore, the quantitative proteomes of neurons and the BBB (or proteotypes) after human brain ischemia presented here contribute to increasing the knowledge regarding the molecular mechanisms of ischemic stroke pathology and highlight new proteins that might represent putative biomarkers of brain ischemia or therapeutic targets.
Collapse
Affiliation(s)
- Teresa García-Berrocoso
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Víctor Llombart
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Colàs-Campàs
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alexandre Hainard
- §Proteomics Core Facility, Faculty of medicine, University Medical Center, University of Geneva, Geneva, Switzerland
| | - Virginie Licker
- ¶Neuroproteomics Group, Human protein sciences department, University Medical Center, University of Geneva, Geneva, Switzerland
| | - Anna Penalba
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Ramiro
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Simats
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alejandro Bustamante
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Martínez-Saez
- ‖Neuropathology, Pathology department, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Canals
- **Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jean-Charles Sanchez
- ‡‡Translational biomarker group, Human protein sciences department, University Medical Center, University of Geneva, Geneva, Switzerland
| | - Joan Montaner
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain;
| |
Collapse
|
39
|
Zhou X, Cheng Y, Zhang R, Li G, Yang B, Zhang S, Wu J. Alpha7 nicotinic acetylcholine receptor agonist promotes retinal ganglion cell function via modulating GABAergic presynaptic activity in a chronic glaucomatous model. Sci Rep 2017; 7:1734. [PMID: 28496108 PMCID: PMC5431927 DOI: 10.1038/s41598-017-02092-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/05/2017] [Indexed: 01/01/2023] Open
Abstract
Alpha-7 nicotinic acetylcholine receptor (α7-nAChR) agonists can prevent glutamate-induced excitotoxicity in cultured retinal ganglion cells (RGCs). However, the neuroprotective effects and the mechanism of action of PNU-282987, an α7-nAChR agonist, in a chronic in vivo rat glaucoma model are poorly understood. We found that elevated intraocular pressure (IOP) downregulated retinal α7-nAChR expression. Electroretinography revealed that the amplitude of the photopic negative response (PhNR) decreased in parallel with the loss of RGCs caused by elevated IOP. PNU-282987 enhanced RGC viability and function and decreased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive signals in RGCs. Patch-clamp recordings revealed differences in the baseline frequencies and decay times of the miniature GABAergic inhibitory postsynaptic currents (mIPSCs) of RGCs between control and glaucomatous retinal slices. The results of western blotting and immunostaining showed that glutamic acid decarboxylase 65/67 and GABA deficits persisted in glaucomatous retinas and that these deficits were reversed by PNU-282987. Patch-clamp recordings also showed that PNU-282987 significantly increased the frequency and amplitude of the GABAergic mIPSCs of RGCs. The protective effects of PNU-292987 were blocked by intravitreal administration of selective GABAA receptor antagonists. The modulation of GABAergic synaptic transmission by PNU-282987 causes de-excitation of ganglion cell circuits and suppresses excitotoxic processes.
Collapse
Affiliation(s)
- Xujiao Zhou
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200032, China.,Key Laboratory of Myopia, Ministry of Health, Shanghai, 200032, China
| | - Yun Cheng
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rong Zhang
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Gang Li
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Boqi Yang
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shenghai Zhang
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jihong Wu
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200032, China. .,Key Laboratory of Myopia, Ministry of Health, Shanghai, 200032, China.
| |
Collapse
|
40
|
The endocannabinoid system, a novel and key participant in acupuncture's multiple beneficial effects. Neurosci Biobehav Rev 2017; 77:340-357. [PMID: 28412017 DOI: 10.1016/j.neubiorev.2017.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/22/2022]
Abstract
Acupuncture and its modified forms have been used to treat multiple medical conditions, but whether the diverse effects of acupuncture are intrinsically linked at the cellular and molecular level and how they might be connected have yet to be determined. Recently, an emerging role for the endocannabinoid system (ECS) in the regulation of a variety of physiological/pathological conditions has been identified. Overlap between the biological and therapeutic effects induced by ECS activation and acupuncture has facilitated investigations into the participation of ECS in the acupuncture-induced beneficial effects, which have shed light on the idea that the ECS may be a primary mediator and regulatory factor of acupuncture's beneficial effects. This review seeks to provide a comprehensive summary of the existing literature concerning the role of endocannabinoid signaling in the various effects of acupuncture, and suggests a novel notion that acupuncture may restore homeostasis under different pathological conditions by regulating similar networks of signaling pathways, resulting in the activation of different reaction cascades in specific tissues in response to pathological insults.
Collapse
|
41
|
Wellendorph P, Jacobsen J, Skovgaard-Petersen J, Jurik A, Vogensen SB, Ecker G, Schousboe A, Krogsgaard-Larsen P, Clausen RP. γ-Aminobutyric Acid and Glycine Neurotransmitter Transporters. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527679430.ch4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Petrine Wellendorph
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Julie Jacobsen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Jonas Skovgaard-Petersen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Andreas Jurik
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14, A-1090 Vienna Austria
| | - Stine B. Vogensen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Gerhard Ecker
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14, A-1090 Vienna Austria
| | - Arne Schousboe
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Povl Krogsgaard-Larsen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Rasmus P. Clausen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| |
Collapse
|
42
|
Glial GABA Transporters as Modulators of Inhibitory Signalling in Epilepsy and Stroke. ADVANCES IN NEUROBIOLOGY 2017; 16:137-167. [PMID: 28828609 DOI: 10.1007/978-3-319-55769-4_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Imbalances in GABA-mediated tonic inhibition are involved in several pathophysiological conditions. A classical way of controlling tonic inhibition is through pharmacological intervention with extrasynaptic GABAA receptors that sense ambient GABA and mediate a persistent GABAergic conductance. An increase in tonic inhibition may, however, also be obtained indirectly by inhibiting glial GABA transporters (GATs). These are sodium-coupled membrane transport proteins that normally act to terminate GABA neurotransmitter action by taking up GABA into surrounding astrocytes. The aim of the review is to provide an overview of glial GATs in regulating tonic inhibition, especially in epilepsy and stroke. This entails a comprehensive summary of changes known to occur in GAT expression levels and signalling following epileptic and ischemic insults. Further, we discuss the accumulating pharmacological evidence for targeting GATs in these diseases.
Collapse
|
43
|
MacKenzie EM, Song MS, Dursun SM, Tomlinson S, Todd KG, Baker GB. Phenelzine: An Old Drug That May Hold Clues to The Development of New Neuroprotective Agents. ACTA ACUST UNITED AC 2016. [DOI: 10.1080/10177833.2010.11790656] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Erin M. MacKenzie
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Mee-Sook Song
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Serdar M. Dursun
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Sara Tomlinson
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Kathryn G. Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Glen B. Baker
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
44
|
The Impact of Cortical Lesions on Thalamo-Cortical Network Dynamics after Acute Ischaemic Stroke: A Combined Experimental and Theoretical Study. PLoS Comput Biol 2016; 12:e1005048. [PMID: 27509209 PMCID: PMC4979968 DOI: 10.1371/journal.pcbi.1005048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/05/2016] [Indexed: 01/25/2023] Open
Abstract
The neocortex and thalamus provide a core substrate for perception, cognition, and action, and are interconnected through different direct and indirect pathways that maintain specific dynamics associated with functional states including wakefulness and sleep. It has been shown that a lack of excitation, or enhanced subcortical inhibition, can disrupt this system and drive thalamic nuclei into an attractor state of low-frequency bursting and further entrainment of thalamo-cortical circuits, also called thalamo-cortical dysrhythmia (TCD). The question remains however whether similar TCD-like phenomena can arise with a cortical origin. For instance, in stroke, a cortical lesion could disrupt thalamo-cortical interactions through an attenuation of the excitatory drive onto the thalamus, creating an imbalance between excitation and inhibition that can lead to a state of TCD. Here we tested this hypothesis by comparing the resting-state EEG recordings of acute ischaemic stroke patients (N = 21) with those of healthy, age-matched control-subjects (N = 17). We observed that these patients displayed the hallmarks of TCD: a characteristic downward shift of dominant α-peaks in the EEG power spectra, together with increased power over the lower frequencies (δ and θ-range). Contrary to general observations in TCD, the patients also displayed a broad reduction in β-band activity. In order to explain the genesis of this stroke-induced TCD, we developed a biologically constrained model of a general thalamo-cortical module, allowing us to identify the specific cellular and network mechanisms involved. Our model showed that a lesion in the cortical component leads to sustained cell membrane hyperpolarization in the corresponding thalamic relay neurons, that in turn leads to the de-inactivation of voltage-gated T-type Ca2+-channels, switching neurons from tonic spiking to a pathological bursting regime. This thalamic bursting synchronises activity on a population level through divergent intrathalamic circuits, and entrains thalamo-cortical pathways by means of propagating low-frequency oscillations beyond the restricted region of the lesion. Hence, pathological stroke-induced thalamo-cortical dynamics can be the source of diaschisis, and account for the dissociation between lesion location and non-specific symptoms of stroke such as neuropathic pain and hemispatial neglect. The thalamus is involved in the relay and processing of most sensory information, and provides an interface between subcortical structures and the neocortex. However, disruptions in the subcortical communication with the thalamus are known to lead to thalamo-cortical dysrhythmia (TCD), which is linked to symptoms in a range of illnesses including Parkinson’s disease, neurogenic pain syndrome and tinnitus. Thus far, TCD has solely been interpreted in terms of changes within subcortical pathways, but here we investigate how cortical disturbances (i.e., ischaemic stroke) may affect thalamic function in a similar manner. We do so by analysing the electroencephalogram (EEG) of stroke patients with a cortical lesion, and show that their EEG power spectra display the characteristic features of TCD. We subsequently built a detailed spiking model of thalamo-cortical circuits to identify the local cellular, circuit, and network properties and dynamics that lead to the development of this stroke-induced TCD. Together, our results shed light on less-understood symptoms of stroke such as neuropathic pain and hemispatial neglect, help inform future brain monitoring and diagnostics post-stroke, and suggest potential new treatments for stroke and related neurological conditions.
Collapse
|
45
|
Curcio M, Salazar IL, Mele M, Canzoniero LMT, Duarte CB. Calpains and neuronal damage in the ischemic brain: The swiss knife in synaptic injury. Prog Neurobiol 2016; 143:1-35. [PMID: 27283248 DOI: 10.1016/j.pneurobio.2016.06.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022]
Abstract
The excessive extracellular accumulation of glutamate in the ischemic brain leads to an overactivation of glutamate receptors with consequent excitotoxic neuronal death. Neuronal demise is largely due to a sustained activation of NMDA receptors for glutamate, with a consequent increase in the intracellular Ca(2+) concentration and activation of calcium- dependent mechanisms. Calpains are a group of Ca(2+)-dependent proteases that truncate specific proteins, and some of the cleavage products remain in the cell, although with a distinct function. Numerous studies have shown pre- and post-synaptic effects of calpains on glutamatergic and GABAergic synapses, targeting membrane- associated proteins as well as intracellular proteins. The resulting changes in the presynaptic proteome alter neurotransmitter release, while the cleavage of postsynaptic proteins affects directly or indirectly the activity of neurotransmitter receptors and downstream mechanisms. These alterations also disturb the balance between excitatory and inhibitory neurotransmission in the brain, with an impact in neuronal demise. In this review we discuss the evidence pointing to a role for calpains in the dysregulation of excitatory and inhibitory synapses in brain ischemia, at the pre- and post-synaptic levels, as well as the functional consequences. Although targeting calpain-dependent mechanisms may constitute a good therapeutic approach for stroke, specific strategies should be developed to avoid non-specific effects given the important regulatory role played by these proteases under normal physiological conditions.
Collapse
Affiliation(s)
- Michele Curcio
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ivan L Salazar
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
| | - Miranda Mele
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal.
| |
Collapse
|
46
|
Endogenous and Synthetic Cannabinoids as Therapeutics in Retinal Disease. Neural Plast 2016; 2016:8373020. [PMID: 26881135 PMCID: PMC4736800 DOI: 10.1155/2016/8373020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/16/2015] [Accepted: 12/01/2015] [Indexed: 11/25/2022] Open
Abstract
The functional significance of cannabinoids in ocular physiology and disease has been reported some decades ago. In the early 1970s, subjects who smoked Cannabis sativa developed lower intraocular pressure (IOP). This led to the isolation of phytocannabinoids from this plant and the study of their therapeutic effects in glaucoma. The main treatment of this disease to date involves the administration of drugs mediating either the decrease of aqueous humour synthesis or the increase of its outflow and thus reduces IOP. However, the reduction of IOP is not sufficient to prevent visual field loss. Retinal diseases, such as glaucoma and diabetic retinopathy, have been defined as neurodegenerative diseases and characterized by ischemia-induced excitotoxicity and loss of retinal neurons. Therefore, new therapeutic strategies must be applied in order to target retinal cell death, reduction of visual acuity, and blindness. The aim of the present review is to address the neuroprotective and therapeutic potential of cannabinoids in retinal disease.
Collapse
|
47
|
Gonzales NR, Grotta JC. Pharmacologic Modification of Acute Cerebral Ischemia. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00055-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
48
|
|
49
|
Hiu T, Farzampour Z, Paz JT, Wang EHJ, Badgely C, Olson A, Micheva KD, Wang G, Lemmens R, Tran KV, Nishiyama Y, Liang X, Hamilton SA, O'Rourke N, Smith SJ, Huguenard JR, Bliss TM, Steinberg GK. Enhanced phasic GABA inhibition during the repair phase of stroke: a novel therapeutic target. Brain 2015; 139:468-80. [PMID: 26685158 PMCID: PMC4805083 DOI: 10.1093/brain/awv360] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/20/2015] [Indexed: 11/13/2022] Open
Abstract
Ischaemic stroke is the leading cause of severe long-term disability yet lacks drug therapies that promote the repair phase of recovery. This repair phase of stroke occurs days to months after stroke onset and involves brain remapping and plasticity within the peri-infarct zone. Elucidating mechanisms that promote this plasticity is critical for the development of new therapeutics with a broad treatment window. Inhibiting tonic (extrasynaptic) GABA signalling during the repair phase was reported to enhance functional recovery in mice suggesting that GABA plays an important function in modulating brain repair. While tonic GABA appears to suppress brain repair after stroke, less is known about the role of phasic (synaptic) GABA during the repair phase. We observed an increase in postsynaptic phasic GABA signalling in mice within the peri-infarct cortex specific to layer 5; we found increased numbers of α1 receptor subunit-containing GABAergic synapses detected using array tomography, and an associated increased efficacy of spontaneous and miniature inhibitory postsynaptic currents in pyramidal neurons. Furthermore, we demonstrate that enhancing phasic GABA signalling using zolpidem, a Food and Drug Administration (FDA)-approved GABA-positive allosteric modulator, during the repair phase improved behavioural recovery. These data identify potentiation of phasic GABA signalling as a novel therapeutic strategy, indicate zolpidem’s potential to improve recovery, and underscore the necessity to distinguish the role of tonic and phasic GABA signalling in stroke recovery.
Collapse
Affiliation(s)
- Takeshi Hiu
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Zoya Farzampour
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Jeanne T Paz
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA 4 Gladstone Institute of Neurological Disease and University of California, San Francisco San Francisco, CA USA
| | - Eric Hou Jen Wang
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Corrine Badgely
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Andrew Olson
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Kristina D Micheva
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Gordon Wang
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Robin Lemmens
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA 6 KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology; VIB - Vesalius Research Center University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium
| | - Kevin V Tran
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Yasuhiro Nishiyama
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Xibin Liang
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Scott A Hamilton
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Nancy O'Rourke
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Stephen J Smith
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - John R Huguenard
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Tonya M Bliss
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Gary K Steinberg
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| |
Collapse
|
50
|
Levetiracetam Prevents Perforin Mediated Neuronal Injury Induced by Acute Cerebral Ischemia Reperfusion. Mol Neurobiol 2015; 53:5480-91. [PMID: 26454821 DOI: 10.1007/s12035-015-9467-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022]
Abstract
The purpose of this study is to explore the neuroprotection mechanism of levetiracetam (LEV) with acute focal cerebral ischemia-reperfusion (I/P) mouse. The cerebral artery I/P animal model was prepared with a middle artery cerebral occlusion method. For drug intervention, mice were intraperitoneally injected with LEV with a dose of either 15 or 150 mg/kg. Neuronal injury was evaluated by measuring the infarct area, apoptosis ratio, and observation of blood-brain barrier ultrastructure with transmission electron microscope. CD8(+) antibody and perforin antibody were used to make cross-reference screen through flow cytometry to determine a perforin-positive rate in CD8(+) T lymphocytes (PFN + %). Injection of LEV can reduce infarct area, apoptosis ratio, and blood-brain barrier damage 24 h later after acute I/P in WT mice. In vitro, perforin can lower hippocampal neuron viability. In vivo, removing perforin can relieve neuronal injury. High dose injection of LEV (150 mg/kg) can inhibit perforin secreting from CD8(+)T lymphocytes. In addition, LEV can still protect neurons with perforin knockout mice. Therefore, our results suggested that LEV may contribute to neuron protection after cerebral ischemia reperfusion. The possible mechanism may be related with perforin release. However, we cannot roll out other mechanisms.
Collapse
|