1
|
Celine G, Thomas M. Temporal characterisation and electrophysiological implications of TBI-induced serine/threonine kinase activity in mouse cortex. Cell Mol Life Sci 2025; 82:102. [PMID: 40045019 PMCID: PMC11883073 DOI: 10.1007/s00018-025-05638-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/05/2025] [Accepted: 02/23/2025] [Indexed: 03/09/2025]
Abstract
Traumatic brain injury (TBI) remains the leading cause of death and disability worldwide with no existing effective treatment. The early phase after TBI induction triggers numerous molecular cascades to regulate adaptive processes and cortical network activity. Kinases play a particularly prominent role in modifying peptide substrates, which include ion channels, receptors, transcription factors and inflammatory mediators. This study aimed to better understand the post-injury serine/threonine kinome; (1) Which kinases conduct phosphorylation-induced alterations of target peptides following unilateral TBI in mouse cortex? (2) How do these kinases effectuate pathological network hyperexcitability, which has detrimental long-term outcomes? We used a serine/threonine kinase assay at 4 h, 24 h and 72 h post-TBI to identify hyper-/hypo-active/phosphorylated kinases and peptides in the ipsilateral and contralateral cortical hemispheres relative to sham-operated controls. We pharmacologically mimicked the changes seen in ERK1/2 and PKC kinase activity, and using microelectrode array recordings we explored their significant electrophysiological implications on spontaneous and evoked cortical activity. We then used these findings to manipulate key kinase activity changes at 24 h post-TBI to rescue the hyperexcitability that is seen in the contralateral cortical network at this timepoint back to sham level. The contribution of specific downstream peptide target channel/receptor subunits was also shown. We conclude that volatile kinase activity has potent implications on cortical network activity after the injury and that these kinases and/or their peptide substrates should be more seriously considered as therapeutic targets for the clinical treatment of TBI.
Collapse
Affiliation(s)
- Gallagher Celine
- Institute of Physiology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Mittmann Thomas
- Institute of Physiology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
2
|
Chen X, Feng Y, Quinn RJ, Pountney DL, Richardson DR, Mellick GD, Ma L. Potassium Channels in Parkinson's Disease: Potential Roles in Its Pathogenesis and Innovative Molecular Targets for Treatment. Pharmacol Rev 2023; 75:758-788. [PMID: 36918260 DOI: 10.1124/pharmrev.122.000743] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/05/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the midbrain. The loss of neurons results in a subsequent reduction of dopamine in the striatum, which underlies the core motor symptoms of PD. To date, there are no effective treatments to stop, slow, or reverse the pathologic progression of dopaminergic neurodegeneration. This unfortunate predicament is because of the current early stages in understanding the biologic targets and pathways involved in PD pathogenesis. Ion channels have become emerging targets for new therapeutic development for PD due to their essential roles in neuronal function and neuroinflammation. Potassium channels are the most prominent ion channel family and have been shown to be critically important in PD pathology because of their roles in modulating neuronal excitability, neurotransmitter release, synaptic transmission, and neuroinflammation. In this review, members of the subfamilies of voltage-gated K+ channels, inward rectifying K+ channels, and Ca2+-activated K+ channels are described. Evidence of the role of these channels in PD etiology is discussed together with the latest views on related pathologic mechanisms and their potential as biologic targets for developing neuroprotective drugs for PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is the second most common neurodegenerative disorder, featuring progressive degeneration of dopaminergic neurons in the midbrain. It is a multifactorial disease involving multiple risk factors and complex pathobiological mechanisms. Mounting evidence suggests that ion channels play vital roles in the pathogenesis and progression of PD by regulating neuronal excitability and immune cell function. Therefore, they have become "hot" biological targets for PD, as demonstrated by multiple clinical trials of drug candidates targeting ion channels for PD therapy.
Collapse
Affiliation(s)
- Xiaoyi Chen
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Yunjiang Feng
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Ronald J Quinn
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Dean L Pountney
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Des R Richardson
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - George D Mellick
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Linlin Ma
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| |
Collapse
|
3
|
Sallam AA, Ahmed MM, El-Magd MA, Magdy A, Ghamry HI, Alshahrani MY, Abou El-Fotoh MF. Quercetin-Ameliorated, Multi-Walled Carbon Nanotubes-Induced Immunotoxic, Inflammatory, and Oxidative Effects in Mice. Molecules 2022; 27:2117. [PMID: 35408516 PMCID: PMC9000348 DOI: 10.3390/molecules27072117] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 12/28/2022] Open
Abstract
The expanding uses of carbon nanotubes (CNTs) in industry and medicine have raised concerns about their toxicity on human and animal health. CNTs, including multi-walled nanotubes (MWCNTs), have been reported to induce immunotoxic, inflammatory, and oxidative effects. Quercetin is a natural flavonoid present in many vegetables and fruits and has immunomodulatory, anti-inflammatory, and antioxidant properties. Herein, we investigated the protective effects of quercetin on pristine MWCNTs-induced immunotoxicity in mice. In comparison with two doses of MWCNTs, high doses [0.5 mg/kg body weight (BW), once intraperitoneally (IP)] caused higher immunotoxic, inflammatory, and oxidative effects than low doses (0.25 mg/kg BW, once IP). Administration of quercetin (30 mg/kg BW, IP for 2 weeks) relieved these deleterious effects as evidenced by (1) reduced spleen weight, (2) increased number of total leukocytes, lymphocytes, and neutrophils, (3) elevated serum levels of IgM, IgG, and IgA, (4) decreased lipid peroxide malondialdehyde levels and increased levels of antioxidant markers reduced glutathione, superoxide dismutase, and catalase in the spleen, (5) decreased concentrations and mRNA levels of inflammatory markers tumor necrosis factor-alpha (TNFα), interleukin 1 beta (IL1ß), and IL6 in the spleen, (6) downregulated expression of immunomodulatory genes transforming growth factor-beta (TGFß), cyclooxygenase2 (COX2), and IL10, and (7) regenerative histological changes as indicated by decreased mononuclear cell infiltration, minimized degenerative changes and restored lymphocytes depletion in the spleen. These results infer that quercetin can ameliorate MWCNTs-induced immunotoxic, inflammatory, and oxidative effects.
Collapse
Affiliation(s)
- Amira A. Sallam
- Department of Forensic Medicine & Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; (A.A.S.); (M.M.A.); (M.F.A.E.-F.)
| | - Mona M. Ahmed
- Department of Forensic Medicine & Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; (A.A.S.); (M.M.A.); (M.F.A.E.-F.)
| | - Mohammed A. El-Magd
- Anatomy Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Ahmed Magdy
- Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Heba I. Ghamry
- Department of Home Economics, College of Home Economics, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Mohammad Y. Alshahrani
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 9088, Abha 61413, Saudi Arabia
| | - Magdy F. Abou El-Fotoh
- Department of Forensic Medicine & Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; (A.A.S.); (M.M.A.); (M.F.A.E.-F.)
| |
Collapse
|
4
|
Senik MH, Abu IF, Fadhullah W. Analysis of K ATP Channels Opening Probability of Hippocampus Cells Treated with Kainic Acid. Malays J Med Sci 2021; 28:15-26. [PMID: 33679216 PMCID: PMC7909348 DOI: 10.21315/mjms2021.28.1.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/12/2020] [Indexed: 01/14/2023] Open
Abstract
Background Kainic acid (KA)-induced seizures may be a valuable tool in the assessment of anti-epileptic drug efficacy in complex partial seizures. This study investigated the effects of KA on ATP-sensitive K+ (KATP) channels opening probability (NPo), which plays a crucial role in neuronal activities. Methods For the optimisation and validation protocol, β-cells were plated onto 35 mm plastic petri dishes and maintained in RPMI-1640 media supplemented with 10 mM glucose, 10% FCS and 25 mM of N-2-hydroxyethylpiperazine-N-ethanesulfonic acid (HEPES). The treatment effects of 10 mM glucose and 30 μM fluoxetine on KATP channels NPo of β-cells were assessed via cell-attached patch-clamp recordings. For hippocampus cell experiments, hippocampi were harvested from day 17 of maternal Lister-hooded rat foetus, and then transferred to a Ca2+ and Mg2+-free HEPES-buffered Hank's salt solution (HHSS). The dissociated cells were cultured and plated onto a 25 mm round cover glasses coated with poly-d-lysine (0.1 mg/mL) in a petri dish. The KATP channels NPo of hippocampus cells when perfused with 1 mM and 10 mM of KA were determined. Results NPo of β-cells showed significant decreasing patterns (P < 0.001) when treated with 10 mM glucose 0.048 (0.027) as well as 30 μM fluoxetine 0.190 (0.141) as compared to basal counterpart. In hippocampus cell experiment, a significant increase (P < 0.001) in mean NPo 2.148 (0.175) of neurons when applied with 1 mM of KA as compared to basal was observed. Conclusion The two concentrations of KA used in the study exerted contrasting effects toward the mean of NPo. It is hypothesised that KA at lower concentration (1 mM) opens more KATP channels, leading to hyperpolarisation of the neurons, which may prevent neuronal hyper excitability. No effect was shown in 10 mM KA treatment, suggesting that only lower than 10 mM KA produced significant changes in KATP channels. This implies further validation of KA concentration to be used in the future.
Collapse
Affiliation(s)
- Mohd Harizal Senik
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,School of Life Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Izuddin Fahmy Abu
- Institute of Medical Science Technology, Universiti Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Widad Fadhullah
- School of Industrial Technology, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| |
Collapse
|
5
|
Abouzed TK, Beltagy EER, Kahilo KA, Ibrahim WM. Molecular changes associated with the anticancer effect of sulforaphane against Ehrlich solid tumour in mice. J Biochem Mol Toxicol 2020; 35:e22655. [PMID: 33094879 DOI: 10.1002/jbt.22655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 10/04/2020] [Accepted: 10/07/2020] [Indexed: 02/05/2023]
Abstract
The anticancer effect of sulforaphane (SFN) is mediated by several signalling pathways. However, little is known regarding the underlying mechanism in Ehrlich solid tumours (ESTs) in mice. This study was conducted to determine molecular changes associated with the anticancer effect of SFN and to compare its preventive (cotreatment) and therapeutic (posttreatment) effects. Ehrlich (murine mammary adenocarcinoma) solid tumour was selected and changes in the gene expression were determined in tumour tissues by the real-time polymerase chain reaction. The results showed that SFN increased the expression of the oxidative stress gene NrF2 and its downstream targets (HO1 and CAT). Conversely, SFN administration decreased the expression of the epigenesis-related genes (HDAC1 and DNMT1) and inflammation-related genes (TNFa, NFkB and Cox2). Overall, SFN cotreatment presented notable molecular changes than the posttreatment strategy. These data suggest that molecular changes associated with the anticancer effects of SFN against EST involved induction of oxidative stress, inhibition of inflammation and epigenetic modifications.
Collapse
Affiliation(s)
- Tarek K Abouzed
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Esraa-Elden R Beltagy
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt.,Department of Clinical Pathology and Clinical Chemistry, Kafrelsheikh University Hospital, Kafr El-Sheikh, Egypt
| | - Khaled A Kahilo
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Wafaa M Ibrahim
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
6
|
Williams G, Gatt A, Clarke E, Corcoran J, Doherty P, Chambers D, Ballard C. Drug repurposing for Alzheimer's disease based on transcriptional profiling of human iPSC-derived cortical neurons. Transl Psychiatry 2019; 9:220. [PMID: 31492831 PMCID: PMC6731247 DOI: 10.1038/s41398-019-0555-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/21/2019] [Accepted: 07/17/2019] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease is a complex disorder encompassing multiple pathological features with associated genetic and molecular culprits. However, target-based therapeutic strategies have so far proved ineffective. The aim of this study is to develop a methodology harnessing the transcriptional changes associated with Alzheimer's disease to develop a high content quantitative disease phenotype that can be used to repurpose existing drugs. Firstly, the Alzheimer's disease gene expression landscape covering severe disease stage, early pathology progression, cognitive decline and animal models of the disease has been defined and used to select a set of 153 drugs tending to oppose disease-associated changes in the context of immortalised human cancer cell lines. The selected compounds have then been assayed in the more biologically relevant setting of iPSC-derived cortical neuron cultures. It is shown that 51 of the drugs drive expression changes consistently opposite to those seen in Alzheimer's disease. It is hoped that the iPSC profiles will serve as a useful resource for drug repositioning within the context of neurodegenerative disease and potentially aid in generating novel multi-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Gareth Williams
- Wolfson Centre for Age-Related Diseases, King's College London, London Bridge, London, SE1 1UL, UK.
| | - Ariana Gatt
- Wolfson Centre for Age-Related Diseases, King's College London, London Bridge, London, SE1 1UL, UK
| | - Earl Clarke
- Wolfson Centre for Age-Related Diseases, King's College London, London Bridge, London, SE1 1UL, UK
| | - Jonathan Corcoran
- Wolfson Centre for Age-Related Diseases, King's College London, London Bridge, London, SE1 1UL, UK
| | - Patrick Doherty
- Wolfson Centre for Age-Related Diseases, King's College London, London Bridge, London, SE1 1UL, UK
| | - David Chambers
- Wolfson Centre for Age-Related Diseases, King's College London, London Bridge, London, SE1 1UL, UK
| | - Clive Ballard
- College of Medicine and Health, University of Exeter, Exeter, EX1 2LU, UK
| |
Collapse
|
7
|
Abu Gazia M, El-Magd MA. Effect of pristine and functionalized multiwalled carbon nanotubes on rat renal cortex. Acta Histochem 2019; 121:207-217. [PMID: 30591315 DOI: 10.1016/j.acthis.2018.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/26/2022]
Abstract
The increasing application of carbon nanotubes (CNTs) within environmental, occupational and consumer settings has raised concerns regarding their biosafety and adverse effects on human health. The present study was designed to investigate the possible adverse effect of pristine and functionalized (amylated and polyethelene glycol coated) multi-walled (MW) CNTs on rat kidney with special concern to the histological alterations and the associated oxidative stress, apoptosis and inflammation. Healthy male albino rats (n = 40) were randomly divided into 4 groups: group I (control), group II (pristine MWCNTs), group III (amylated MWCNTs) and group IV [polyethelene glycol (PEG)-coated MWCNTs]. Animals of groups II, III and IV received a single dose of 1 mg/kg body weight of MWCNTs via intra-tracheal (IT) instillation at the beginning of the experiment and all rats were sacrificed after 30 days. Rats in groups II and III showed, nearly similar, renal tissue damage (evidenced by thin collapsed glomeruli, packed mesangial and endothelial cells as well as edematous hemorrhagic glomeruli with apoptotic changes) and functional disruptions (indicated by high serum levels of urea and creatinine) probably through induction of oxidative stress [revealed by high level of the lipid peroxidation marker malondialdehyde (MDA) and lower levels of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx)], apoptosis (indicated by high caspase 3 activity), and inflammation (evidenced by high level of IL1β). However, PEG-coated MWCNTs-treated group (group IV) showed nearly normal renal structure and function. It could be concluded that pristine and functionalized amylated MWCNTs have nephrotoxic effect, while PEG-coated MWCNTs had lowest, or none, toxic effects making them safer for therapy and diagnosis of a variety of diseases.
Collapse
|
8
|
Xin H, Cui Y, An Z, Yang Q, Zou X, Yu N. Attenuated glutamate induced ROS production by antioxidative compounds in neural cell lines. RSC Adv 2019; 9:34735-34743. [PMID: 35530670 PMCID: PMC9074000 DOI: 10.1039/c9ra03848e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
Glutamate is an excitatory neurotransmitter involved in neural function. Excess accumulation of intercellular glutamate leads to increasing concentration of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in neuronal cells. In this study, we investigated the antioxidant activity of several typical superior compounds among four neuronal cells, and determined the scavenging activity of free radicals. The in vivo assay was also carried out to compare the protective effect of glutamate-induced cell damage. Hierarchical clustering analysis was used to identify the common properties. Glutamate induced neurotoxicity and ROS production, suggesting glutamate cytotoxicity was related to oxidative stress and widely exists in different cell lines. Those screening compounds exhibited strong antioxidant ability, but low cytotoxicity to neuronal cells, acting as agents against neurodegenerative diseases. Finally, a hierarchical clustering analysis assay indicated that hyperoside and rutin hydrate are the most effective compounds for attenuating intercellular ROS levels. The results suggested the activity more or less relies on structure, rather than residues. These data generate new supporting ideas to remove intracellular ROS and the identified compounds serve as potential therapeutic agents in multiple neurological diseases. Glutamate is an excitatory neurotransmitter involved in neural function.![]()
Collapse
Affiliation(s)
- Haolin Xin
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Ying Cui
- Tianjin University of Traditional Chinese Medicine
- Tianjin
- China
| | - Zhongping An
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Qian Yang
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Xuan Zou
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| | - Ning Yu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases
- Department of Neurology
- Nankai University
- Huanhu Hospital
- Tianjin
| |
Collapse
|
9
|
Vidal-Taboada JM, Pugliese M, Salvadó M, Gámez J, Mahy N, Rodríguez MJ. K ATP Channel Expression and Genetic Polymorphisms Associated with Progression and Survival in Amyotrophic Lateral Sclerosis. Mol Neurobiol 2018; 55:7962-7972. [PMID: 29492846 DOI: 10.1007/s12035-018-0970-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/16/2018] [Indexed: 02/06/2023]
Abstract
The ATP-sensitive potassium (KATP) channel directly regulates the microglia-mediated inflammatory response following CNS injury. To determine the putative role of the KATP channel in amyotrophic lateral sclerosis (ALS) pathology, we investigated whether ALS induces changes in KATP channel expression in the spinal cord and motor cortex. We also characterized new functional variants of human ABCC8, ABCC9, KCNJ8, and KCNJ11 genes encoding for the KATP channel and analyzed their association with ALS risk, rate of progression, and survival in a Spanish ALS cohort. The expression of ABCC8 and KCNJ8 genes was enhanced in the spinal cord of ALS samples, and KCNJ11 increased in motor cortex of ALS samples, as determined by real-time polymerase chain reaction. We then sequenced the exons and regulatory regions of KATP channel genes from a subset of 28 ALS patients and identified 50 new genetic variants. For the case-control association analysis, we genotyped five selected polymorphisms with predicted functional relevance in 185 Spanish ALS (134 spinal ALS and 51 bulbar ALS) patients and 493 controls. We found that bulbar ALS patients presenting the G/G genotype of the rs4148646 variant of ABCC8 and the T/T genotype of the rs5219 variant of KCNJ11 survived longer than other ALS patients presenting other genotypes. Also, the C/C genotype of the rs4148642 variant of ABCC8 and the T/C genotype of the rs148416760 variant of ABCC9 modified the progression rate in spinal ALS patients. Our results suggest that the KATP channel plays a role in the pathophysiological mechanisms of ALS.
Collapse
Affiliation(s)
- José M Vidal-Taboada
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Marco Pugliese
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Maria Salvadó
- ALS Unit, Department of Neurology, Hospital Universitari Vall d'Hebron - VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Josep Gámez
- ALS Unit, Department of Neurology, Hospital Universitari Vall d'Hebron - VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nicole Mahy
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Manuel J Rodríguez
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain. .,Unitat de Bioquímica i Biologia Molecular, Department of Biomedicina, Facultat de Medicina, UB, c/ Casanova 143, E-08036, Barcelona, Spain.
| |
Collapse
|
10
|
Deryagin OG, Gavrilova SA, Gainutdinov KL, Golubeva AV, Andrianov VV, Yafarova GG, Buravkov SV, Koshelev VB. Molecular Bases of Brain Preconditioning. Front Neurosci 2017; 11:427. [PMID: 28790886 PMCID: PMC5524930 DOI: 10.3389/fnins.2017.00427] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022] Open
Abstract
Preconditioning of the brain induces tolerance to the damaging effects of ischemia and prevents cell death in ischemic penumbra. The development of this phenomenon is mediated by mitochondrial adenosine triphosphate-sensitive potassium (KATP+) channels and nitric oxide signaling (NO). The aim of this study was to investigate the dynamics of molecular changes in mitochondria after ischemic preconditioning (IP) and the effect of pharmacological preconditioning (PhP) with the KATP+-channels opener diazoxide on NO levels after ischemic stroke in rats. Immunofluorescence-histochemistry and laser-confocal microscopy were applied to evaluate the cortical expression of electron transport chain enzymes, mitochondrial KATP+-channels, neuronal and inducible NO-synthases, as well as the dynamics of nitrosylation and nitration of proteins in rats during the early and delayed phases of IP. NO cerebral content was studied with electron paramagnetic resonance (EPR) spectroscopy using spin trapping. We found that 24 h after IP in rats, there is a two-fold decrease in expression of mitochondrial KATP+-channels (p = 0.012) in nervous tissue, a comparable increase in expression of cytochrome c oxidase (p = 0.008), and a decrease in intensity of protein S-nitrosylation and nitration (p = 0.0004 and p = 0.001, respectively). PhP led to a 56% reduction of free NO concentration 72 h after ischemic stroke simulation (p = 0.002). We attribute this result to the restructuring of tissue energy metabolism, namely the provision of increased catalytic sites to mitochondria and the increased elimination of NO, which prevents a decrease in cell sensitivity to oxygen during subsequent periods of severe ischemia.
Collapse
Affiliation(s)
- Oleg G Deryagin
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Svetlana A Gavrilova
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Khalil L Gainutdinov
- Laboratory of Neurorehabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal UniversityKazan, Russia.,Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of SciencesKazan, Russia
| | - Anna V Golubeva
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Vyatcheslav V Andrianov
- Laboratory of Neurorehabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal UniversityKazan, Russia.,Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of SciencesKazan, Russia
| | - Guzel G Yafarova
- Laboratory of Neurorehabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal UniversityKazan, Russia.,Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of SciencesKazan, Russia
| | - Sergey V Buravkov
- Research Laboratory of Cellular Structure and Tissue Imaging Analysis, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| | - Vladimir B Koshelev
- Department of Physiology and General Pathology, Medical Faculty, Lomonosov Moscow State UniversityMoscow, Russia
| |
Collapse
|
11
|
Mao XY, Zhou HH, Li X, Liu ZQ. Huperzine A Alleviates Oxidative Glutamate Toxicity in Hippocampal HT22 Cells via Activating BDNF/TrkB-Dependent PI3K/Akt/mTOR Signaling Pathway. Cell Mol Neurobiol 2016; 36:915-925. [PMID: 26440805 PMCID: PMC11482372 DOI: 10.1007/s10571-015-0276-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 09/28/2015] [Indexed: 12/15/2022]
Abstract
Oxidative glutamate toxicity is involved in diverse neurological disorders including epilepsy and ischemic stroke. Our present work aimed to assess protective effects of huperzine A (HupA) against oxidative glutamate toxicity in a mouse-derived hippocampal HT22 cells and explore its potential mechanisms. Cell survival and cell injury were analyzed by MTT method and LDH release assay, respectively. The production of ROS was measured by detection kits. Protein expressions of BDNF, phosphor-TrkB (p-TrkB), TrkB, phosphor-Akt (p-Akt), Akt, phosphor-mTOR (p-mTOR), mTOR, phosphor-p70s6 (p-p70s6) kinase, p70s6 kinase, Bcl-2, Bax, and β-actin were assayed via Western blot analysis. Enzyme-linked immunosorbent assay was employed to measure the contents of nerve growth factor, brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4). Our findings illustrated 10 μM HupA for 24 h significantly protected HT22 from cellular damage and suppressed the generation of ROS. Additionally, after treating with LY294002 or wortmannin [the selective inhibitors of phosphatidylinositol 3 kinase (PI3K)], HupA dramatically prevented the down-regulations of p-Akt, p-mTOR, and p-p70s6 kinase in HT22 cells under oxidative toxicity. Furthermore, it was observed that the protein levels of BDNF and p-TrkB were evidently enhanced after co-treatment with HupA and glutamate in HT22 cells. The elevations of p-Akt and p-mTOR were abrogated under toxic conditions after blockade of TrkB by TrkB IgG. Cellular apoptosis was significantly suppressed (decreased caspase-3 activity and enhanced Bcl-2 protein level) after HupA treatment. It was concluded that HupA attenuated oxidative glutamate toxicity in murine hippocampal HT22 cells via activating BDNF/TrkB-dependent PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiao-Yuan Mao
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, Hunan, P. R. China.
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, People's Republic of China.
| | - Hong-Hao Zhou
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, Hunan, P. R. China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, People's Republic of China
| | - Xi Li
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, Hunan, P. R. China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, People's Republic of China
| | - Zhao-Qian Liu
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, Hunan, P. R. China.
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, People's Republic of China.
| |
Collapse
|
12
|
Martínez-Moreno M, Batlle M, Ortega FJ, Gimeno-Bayón J, Andrade C, Mahy N, Rodríguez MJ. Diazoxide enhances excitotoxicity-induced neurogenesis and attenuates neurodegeneration in the rat non-neurogenic hippocampus. Neuroscience 2016; 333:229-43. [PMID: 27471195 DOI: 10.1016/j.neuroscience.2016.07.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/13/2022]
Abstract
Diazoxide, a well-known mitochondrial KATP channel opener with neuroprotective effects, has been proposed for the effective and safe treatment of neuroinflammation. To test whether diazoxide affects the neurogenesis associated with excitotoxicity in brain injury, we induced lesions by injecting excitotoxic N-methyl-d-aspartate (NMDA) into the rat hippocampus and analyzed the effects of a daily oral administration of diazoxide on the induced lesion. Specific glial and neuronal staining showed that NMDA elicited a strong glial reaction associated with progressive neuronal loss in the whole hippocampal formation. Doublecortin immunohistochemistry and bromo-deoxyuridine (BrdU)-NeuN double immunohistochemistry revealed that NMDA also induced cell proliferation and neurogenesis in the lesioned non-neurogenic hippocampus. Furthermore, glial fibrillary acidic protein (GFAP)-positive cells in the injured hippocampus expressed transcription factor Sp8 indicating that the excitotoxic lesion elicited the migration of progenitors from the subventricular zone and/or the reprograming of reactive astrocytes. Diazoxide treatment attenuated the NMDA-induced hippocampal injury in rats, as demonstrated by decreases in the size of the lesion, neuronal loss and microglial reaction. Diazoxide also increased the number of BrdU/NeuN double-stained cells and elevated the number of Sp8-positive cells in the lesioned hippocampus. These results indicate a role for KATP channel activation in regulating excitotoxicity-induced neurogenesis in brain injury.
Collapse
Affiliation(s)
- M Martínez-Moreno
- Departament de Biomedicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - M Batlle
- Departament de Biomedicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - F J Ortega
- Departament de Biomedicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - J Gimeno-Bayón
- Departament de Biomedicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - C Andrade
- Departament de Biomedicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - N Mahy
- Departament de Biomedicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - M J Rodríguez
- Departament de Biomedicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
| |
Collapse
|