1
|
Uğuz AC, Okan A, Doğanyiğit Z, Yilmaz S, Ateş Ş, Arikan Söylemez ES, Karabulut S, Kumru AS, Espino J. Evaluation of TRPM2 Channel-Mediated Autophagic Signaling Pathway in Hippocampus and Cortex Tissues of Rat Offspring Following Prenatal Exposure to Elevated Alcohol Levels. ENVIRONMENTAL TOXICOLOGY 2024. [PMID: 39387650 DOI: 10.1002/tox.24427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/05/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
Fetal alcohol syndrome (FAS) can occur because of high amount of alcohol intake during pregnancy and is characterized by both physical and neurological problems. Children diagnosed with FAS have difficulties in learning, memory, and coordination. Hippocampus has a major role in memory and learning. We aimed to determine whether alcohol exposure during pregnancy had any effect on offspring by evaluating learning ability as well as oxidative stress and autophagy in the hippocampus and cortex tissues of litters. Attention was also paid to sex differences. To do so, TRPM2, Beclin1, p62, LC3B, IBA1, parvalbumin, GAD65, and mGluR5 expression levels were evaluated by immunohistochemistry. Lactate dehydrogenase (LDH), and malondialdehyde (MDA) levels, as well as total oxidant (TOS) and total antioxidant (TAS) status were determined by ELISA. Learning experiments were evaluated by the Morris water maze (MWM) test. Our findings demonstrated that IBA1, LC3B, GAD65, and mGluR5 expression levels were higher in female rats of the chronic alcohol exposure (CAE) model. Our IHC results revealed that TRPM2 expression levels were significantly increased in both males and females in the CAE group. Likewise, TAS was lower, and TOS was higher in CAE animals. Moreover, MWM outcomes supported a learning deficiency in CAE litters compared to controls and indicated that female offspring outperformed males in learning experiments. Therefore, our results revealed the detrimental effects of alcohol exposure during pregnancy on autophagy signaling in the hippocampus and cortex tissue of litters, which could affect the learning ability of animals.
Collapse
Affiliation(s)
- Abdülhadi Cihangir Uğuz
- Department of Biophysics, School of Medicine, Karamanoğlu Mehmetbey University, Karaman, Türkiye
| | - Aslı Okan
- Department of Histology and Embryology, School of Medicine, Yozgat Bozok University, Yozgat, Türkiye
| | - Züleyha Doğanyiğit
- Department of Histology and Embryology, School of Medicine, Yozgat Bozok University, Yozgat, Türkiye
| | - Seher Yilmaz
- Department of Anatomy, School of Medicine, Yozgat Bozok University, Yozgat, Türkiye
| | - Şükrü Ateş
- Department of Anatomy, School of Medicine, Yozgat Bozok University, Yozgat, Türkiye
| | - Evrim Suna Arikan Söylemez
- Department of Medical Biology, School of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Türkiye
| | - Sebahattin Karabulut
- Department of Physiology, School of Medicine, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Alper Serhat Kumru
- Department of Pharmacology and Toxicology, School of Veterinary Medicine, Sivas, Türkiye
| | - Javier Espino
- Department of Physiology, Faculty of Science, University of Extremadura, Badajoz, Spain
| |
Collapse
|
2
|
Thiankhaw K, Chattipakorn N, Chattipakorn SC. How calcineurin inhibitors affect cognition. Acta Physiol (Oxf) 2024; 240:e14161. [PMID: 38747643 DOI: 10.1111/apha.14161] [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: 11/18/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
AIMS With a focus on the discrepancy between preclinical and clinical findings, this review will gather comprehensive information about the effects of calcineurin inhibitors (CNI) on cognitive function and related brain pathology from in vitro, in vivo, and clinical studies. We also summarize the potential mechanisms that underlie the pathways related to CNI-induced cognitive impairment. METHODS We systematically searched articles in PubMed using keywords 'calcineurin inhibitor*' and 'cognition' to identify related articles, which the final list pertaining to underlying mechanisms of CNI on cognition. RESULTS Several studies have reported an association between calcineurin and the neuropathology of Alzheimer's disease (AD). AD is the most common neurocognitive disorder associated with amyloid plaques and neurofibrillary tangles in the brain, leading to cognitive impairment. CNI, including tacrolimus and cyclosporin A, are commonly prescribed for patients with transplantation of solid organs such as kidney, liver, or heart, those drugs are currently being used as long-term immunosuppressive therapy. Although preclinical models emphasize the favorable effects of CNI on the restoration of brain pathology due to the impacts of calcineurin on the alleviation of amyloid-beta deposition and tau hyperphosphorylation, or rescuing synaptic and mitochondrial functions, treatment-related neurotoxicity, resulting in cognitive dysfunctions has been observed in clinical settings of patients who received CNI. CONCLUSION Inconsistent results of CNI on cognition from clinical studies have been observed due to impairment of the blood-brain barrier, neuroinflammation mediated by reactive oxygen species, and alteration in mitochondrial fission, and extended research is required to confirm its promising use in cognitive impairment.
Collapse
Affiliation(s)
- Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siripron C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
3
|
Burch AM, Garcia JD, O'Leary H, Haas A, Orfila JE, Tiemeier E, Chalmers N, Smith KR, Quillinan N, Herson PS. TRPM2 and CaMKII Signaling Drives Excessive GABAergic Synaptic Inhibition Following Ischemia. J Neurosci 2024; 44:e1762232024. [PMID: 38565288 PMCID: PMC11079974 DOI: 10.1523/jneurosci.1762-23.2024] [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: 09/22/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Excitotoxicity and the concurrent loss of inhibition are well-defined mechanisms driving acute elevation in excitatory/inhibitory (E/I) balance and neuronal cell death following an ischemic insult to the brain. Despite the high prevalence of long-term disability in survivors of global cerebral ischemia (GCI) as a consequence of cardiac arrest, it remains unclear whether E/I imbalance persists beyond the acute phase and negatively affects functional recovery. We previously demonstrated sustained impairment of long-term potentiation (LTP) in hippocampal CA1 neurons correlating with deficits in learning and memory tasks in a murine model of cardiac arrest/cardiopulmonary resuscitation (CA/CPR). Here, we use CA/CPR and an in vitro ischemia model to elucidate mechanisms by which E/I imbalance contributes to ongoing hippocampal dysfunction in male mice. We reveal increased postsynaptic GABAA receptor (GABAAR) clustering and function in the CA1 region of the hippocampus that reduces the E/I ratio. Importantly, reduced GABAAR clustering observed in the first 24 h rebounds to an elevation of GABAergic clustering by 3 d postischemia. This increase in GABAergic inhibition required activation of the Ca2+-permeable ion channel transient receptor potential melastatin-2 (TRPM2), previously implicated in persistent LTP and memory deficits following CA/CPR. Furthermore, we find Ca2+-signaling, likely downstream of TRPM2 activation, upregulates Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, thereby driving the elevation of postsynaptic inhibitory function. Thus, we propose a novel mechanism by which inhibitory synaptic strength is upregulated in the context of ischemia and identify TRPM2 and CaMKII as potential pharmacological targets to restore perturbed synaptic plasticity and ameliorate cognitive function.
Collapse
Affiliation(s)
- Amelia M Burch
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Joshua D Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Heather O'Leary
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Ami Haas
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - James E Orfila
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - Erika Tiemeier
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Nicholas Chalmers
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Nidia Quillinan
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Paco S Herson
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio 43210
| |
Collapse
|
4
|
Ge C, Wang X, Wang Y, Lei L, Song G, Qian M, Wang S. PKCε inhibition prevents ischemia‑induced dendritic spine impairment in cultured primary neurons. Exp Ther Med 2023; 25:152. [PMID: 36911376 PMCID: PMC9995843 DOI: 10.3892/etm.2023.11851] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Brain ischemia is an independent risk factor for Alzheimer's disease (AD); however, the mechanisms underlining ischemic stroke and AD remain unclear. The present study aimed to investigate the function of the ε isoform of protein kinase C (PKCε) in brain ischemia-induced dendritic spine dysfunction to elucidate how brain ischemia causes AD. In the present study, primary hippocampus and cortical neurons were cultured while an oxygen-glucose deprivation (OGD) model was used to simulate brain ischemia. In the OGD cell model, in vitro kinase activity assay was performed to investigate whether the PKCε kinase activity changed after OGD treatment. Confocal microscopy was performed to investigate whether inhibiting PKCε kinase activity protects dendritic spine morphology and function. G-LISA was used to investigate whether small GTPases worked downstream of PKCε. The results showed that PKCε kinase activity was significantly increased following OGD treatment in primary neurons, leading to dendritic spine dysfunction. Pre-treatment with PKCε-inhibiting peptide, which blocks PKCε activity, significantly rescued dendritic spine function following OGD treatment. Furthermore, PKCε could activate Ras homolog gene family member A (RhoA) as a downstream molecule, which mediated OGD-induced dendritic spine morphology changes and caused dendritic spine dysfunction. In conclusion, the present study demonstrated that the PKCε/RhoA signalling pathway is a novel mechanism mediating brain ischemia-induced dendritic spine dysfunction. Developing therapeutic targets for this pathway may protect against and prevent brain ischemia-induced cognitive impairment and AD.
Collapse
Affiliation(s)
- Chenjie Ge
- Department of Psychiatry, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Xuefeng Wang
- WuXi AppTec Co., Ltd., Shanghai 200131, P.R. China
| | - Yunhong Wang
- WuXi AppTec Co., Ltd., Shanghai 200131, P.R. China
| | - Lilei Lei
- Department of Psychiatry, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Guohua Song
- Department of Psychiatry, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Mincai Qian
- Department of Psychiatry, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Shiliang Wang
- Department of Psychiatry, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
5
|
Involvement of DAAO Overexpression in Delayed Hippocampal Neuronal Death. Cells 2022; 11:cells11223689. [PMID: 36429117 PMCID: PMC9688509 DOI: 10.3390/cells11223689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND D-amino acid oxidase (DAAO) is a flavoenzyme that specifically catalyzes the deamination of many neutral and basic D-amino acids. This study aims to explore the pathological increment of hippocampal DAAO and its potential relationship with delayed hippocampal neuronal death. METHODS Ischemia-reperfusion was induced in mice through middle cerebral artery occlusion (MCAO). Neurological deficit scores and hippocampal neuronal death were assessed in MCAO mice. Immunofluorescent staining was applied to identify activated astrocytes and evaluate DAAO expression. TUNEL and Nissl staining were utilized to identify cell apoptosis of hippocampal neurons. RESULTS Hippocampal astrocytic DAAO was strikingly increased following ischemic stroke, with the greatest increase on day 5 after surgery, followed by the manifestation of neurobehavioral deficits. Astrocytic DAAO was found to be mainly expressed in the hippocampal CA2 region and linked with subsequent specific neural apoptosis. Thus, it is supposed that the activation of astrocytic DAAO in ischemic stroke might contribute to neuronal death. An intravenous, twice-daily administration of 4H-furo[3,2-b]pyrrole-5-carboxylic acid (SUN, 10 mg/kg) markedly relieved behavioral status and delayed hippocampal neuronal death by 38.0% and 41.5%, respectively, compared to the model group treated with saline. In transfected primary astrocytes, DAAO overexpression inhibits cell activity, induces cytotoxicity, and promotes hippocampal neuronal death at least partly by enhancing H2O2 levels with subsequent activation of TRP calcium channels in neurons. CONCLUSIONS Our findings suggest that increased hippocampal DAAO is causally associated with the development of delayed neuronal death after MCAO onset via astrocyte-neuron interactions. Hence, targeting DAAO is a promising therapeutic strategy for the management of neurological disorders.
Collapse
|
6
|
Vaidya B, Kaur H, Thapak P, Sharma SS, Singh JN. Pharmacological Modulation of TRPM2 Channels via PARP Pathway Leads to Neuroprotection in MPTP-induced Parkinson's Disease in Sprague Dawley Rats. Mol Neurobiol 2022; 59:1528-1542. [PMID: 34997907 DOI: 10.1007/s12035-021-02711-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Transient receptor potential melastatin-2 (TRPM2) channels are cation channels activated by oxidative stress and ADP-ribose (ADPR). Role of TRPM2 channels has been postulated in several neurological disorders, but, it has not been explored in animal models of Parkinson's disease (PD). Thus, the role of TRPM2 and its associated poly (ADPR) polymerase (PARP) signaling pathways were investigated in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD rat model using TRPM2 inhibitor, 2-aminoethyl diphenyl borinate (2-APB), and PARP inhibitor, N-(6-Oxo-5,6-dihydrophenanthridin-2-yl)-(N,N-dimethylamino) acetamide hydrochloride (PJ-34). PD was induced by using a bilateral intranigral administration of MPTP in rats, and different parameters were evaluated. An increase in oxidative stress was observed, leading to locomotor and cognitive deficits in the PD rats. PD rats also showed an increased TRPM2 expression in the striatum and mid-brain accompanied by reduced expression of tyrosine hydroxylase (TH) in comparison to sham animals. Intraperitoneal administration of 2-APB and PJ-34 led to an improvement in the locomotor and cognitive deficits in comparison to MPTP-induced PD rats. These improvements were accompanied by a reduction in the levels of oxidative stress and an increase in TH levels in the striatum and mid-brain. In addition, these pharmacological interventions also led to a decrease in the expression of TRPM2 in PD in the striatum and mid-brain. Our results provide a rationale for the development of potent pharmacological agents targeting the TRPM2-PARP pathway to provide therapeutic benefits for the treatment of neurological diseases like PD.
Collapse
Affiliation(s)
- Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Harpinder Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India
| | - Jitendra Narain Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar (Mohali), 160062, Punjab, India.
| |
Collapse
|
7
|
Wang Q, Liu N, Ni YS, Yang JM, Ma L, Lan XB, Wu J, Niu JG, Yu JQ. TRPM2 in ischemic stroke: Structure, molecular mechanisms, and drug intervention. Channels (Austin) 2021; 15:136-154. [PMID: 33455532 PMCID: PMC7833771 DOI: 10.1080/19336950.2020.1870088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 01/14/2023] Open
Abstract
Ischemic stroke has a high lethality rate worldwide, and novel treatments are limited. Calcium overload is considered to be one of the mechanisms of cerebral ischemia. Transient receptor potential melastatin 2 (TRPM2) is a reactive oxygen species (ROS)-sensitive calcium channel. Cerebral ischemia-induced TRPM2 activation triggers abnormal intracellular Ca2+ accumulation and cell death, which in turn causes irreversible brain damage. Thus, TRPM2 has emerged as a new therapeutic target for ischemic stroke. This review provides data on the expression, structure, and function of TRPM2 and illustrates its cellular and molecular mechanisms in ischemic stroke. Natural and synthetic TRPM2 inhibitors (both specific and nonspecific) are also summarized. The three-dimensional protein structure of TRPM2 has been identified, and we speculate that molecular simulation techniques will be essential for developing new drugs that block TRPM2 channels. These insights about TRPM2 may be the key to find potent therapeutic approaches for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qing Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Yuan-Shu Ni
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jia-Mei Yang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Lin Ma
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Xiao-Bing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jing Wu
- Laboratory Animal Center, Ningxia Medical University, Yinchuan, China
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Jian-Qiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
- Ningxia Collaborative Innovation Center of Regional Characteristic Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
8
|
Zhu T, Zhu M, Qiu Y, Wu Z, Huang N, Wan G, Xu J, Song P, Wang S, Yin Y, Li P. Puerarin Alleviates Vascular Cognitive Impairment in Vascular Dementia Rats. Front Behav Neurosci 2021; 15:717008. [PMID: 34720898 PMCID: PMC8554240 DOI: 10.3389/fnbeh.2021.717008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Cerebral ischemia triggers vascular dementia (VD), which is characterized by memory loss, cognitive deficits, and vascular injury in the brain. Puerarin (Pur) represents the major isoflavone glycoside of Radix Puerariae, with verified neuroprotective activity and cardiovascular protective effects. However, whether Pur ameliorates cognitive impairment and vascular injury in rats with permanent occlusion of bilateral common carotid arteries (BCCAO) remains unknown. This work aimed to assess Pur's effects on BCCAO-induced VD and to dissect the underlying mechanisms, especially examining the function of transient receptor potential melastatin-related 2 (TRPM2) in alleviating cognitive deficits and vascular injuries. Rats with BCCAO developed VD. Pur (50, 100, and 150 mg/kg) dose-dependently attenuated the pathological changes, increased synaptic structural plasticity in the dorsal CA1 hippocampal region and decreased oxidative stress, which eventually reduced cognitive impairment and vascular injury in BCCAO rats. Notably, Pur-improved neuronal cell loss, synaptic structural plasticity, and endothelial vasorelaxation function might be mediated by the reactive oxygen species (ROS)-dependent TRPM2/NMDAR pathway, evidenced by decreased levels of ROS, malondialdehyde (MDA), Bax, Bax/Bcl2, and TRPM2, and increased levels of superoxide dismutase (SOD), Bcl2, and NR2A. In conclusion, Pur has therapeutic potential for VD, alleviating neuronal cell apoptosis and vascular injury, which may be related to the ROS-dependent TRPM2/NMDAR pathway.
Collapse
Affiliation(s)
- Tiantian Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Moli Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yue Qiu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Zeqing Wu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Ning Huang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Guangrui Wan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Jian Xu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Ping Song
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shuangxi Wang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yaling Yin
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| |
Collapse
|
9
|
Functional Restoration following Global Cerebral Ischemia in Juvenile Mice following Inhibition of Transient Receptor Potential M2 (TRPM2) Ion Channels. Neural Plast 2021; 2021:8774663. [PMID: 34659399 PMCID: PMC8514917 DOI: 10.1155/2021/8774663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
Hippocampal cell death and cognitive dysfunction are common following global cerebral ischemia across all ages, including children. Most research has focused on preventing neuronal death. Restoration of neuronal function after cell death is an alternative approach (neurorestoration). We previously identified transient receptor potential M2 (TRPM2) ion channels as a potential target for acute neuroprotection and delayed neurorestoration in an adult CA/CPR mouse model. Cardiac arrest/cardiopulmonary resuscitation (CA/CPR) in juvenile (p20-25) mice was used to investigate the role of ion TRPM2 channels in neuroprotection and ischemia-induced synaptic dysfunction in the developing brain. Our novel TRPM2 inhibitor, tatM2NX, did not confer protection against CA1 pyramidal cell death but attenuated synaptic plasticity (long-term plasticity (LTP)) deficits in both sexes. Further, in vivo administration of tatM2NX two weeks after CA/CPR reduced LTP impairments and restored memory function. These data provide evidence that pharmacological synaptic restoration of the surviving hippocampal network can occur independent of neuroprotection via inhibition of TRPM2 channels, providing a novel strategy to improve cognitive recovery in children following cerebral ischemia. Importantly, these data underscore the importance of age-appropriate models in disease research.
Collapse
|
10
|
Cruz Del Angel Y, Orfila JE, Herson PS, Brooks-Kayal A, González MI. Down-regulation of AMPA receptors and long-term potentiation during early epileptogenesis. Epilepsy Behav 2021; 124:108320. [PMID: 34592633 DOI: 10.1016/j.yebeh.2021.108320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Epilepsy is a brain disorder characterized by the occurrence of recurrent spontaneous seizures. Behavioral disorders and altered cognition are frequent comorbidities affecting the quality of life of people with epilepsy. These impairments are undoubtedly multifactorial and the specific mechanisms underlying these comorbidities are largely unknown. Long-lasting alterations in synaptic strength due to changes in expression, phosphorylation, or function of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors (AMPARs) have been associated with alterations in neuronal synaptic plasticity. In particular, alterations in hippocampal long-term potentiation (LTP), a well-accepted model of learning and memory, have been associated with altered cognition in epilepsy. Here, we analyzed the effects of pilocarpine-induced status epilepticus (SE) on AMPARs to determine if alterations in AMPAR signaling might be one of the mechanisms contributing to altered cognition during epilepsy. We found alterations in the phosphorylation and plasma membrane expression of AMPARs. In addition, we detected altered expression of GRIP, a key scaffolding protein involved in the proper distribution of AMPARs at the neuronal cell surface. Interestingly, a functional analysis revealed that these molecular changes are linked to impaired LTP. Together, these observations suggest that seizure-induced alterations in the molecular machinery regulating AMPARs likely impact the neuron's ability to support synaptic plasticity that is required for learning and memory.
Collapse
Affiliation(s)
- Yasmin Cruz Del Angel
- Department of Pediatrics, Division of Neurology and Translational Epilepsy Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - James E Orfila
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paco S Herson
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Amy Brooks-Kayal
- Department of Pediatrics, Division of Neurology and Translational Epilepsy Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Marco I González
- Department of Pediatrics, Division of Neurology and Translational Epilepsy Research Program, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
11
|
Advances in TRP channel drug discovery: from target validation to clinical studies. Nat Rev Drug Discov 2021; 21:41-59. [PMID: 34526696 PMCID: PMC8442523 DOI: 10.1038/s41573-021-00268-4] [Citation(s) in RCA: 212] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/20/2022]
Abstract
Transient receptor potential (TRP) channels are multifunctional signalling molecules with many roles in sensory perception and cellular physiology. Therefore, it is not surprising that TRP channels have been implicated in numerous diseases, including hereditary disorders caused by defects in genes encoding TRP channels (TRP channelopathies). Most TRP channels are located at the cell surface, which makes them generally accessible drug targets. Early drug discovery efforts to target TRP channels focused on pain, but as our knowledge of TRP channels and their role in health and disease has grown, these efforts have expanded into new clinical indications, ranging from respiratory disorders through neurological and psychiatric diseases to diabetes and cancer. In this Review, we discuss recent findings in TRP channel structural biology that can affect both drug development and clinical indications. We also discuss the clinical promise of novel TRP channel modulators, aimed at both established and emerging targets. Last, we address the challenges that these compounds may face in clinical practice, including the need for carefully targeted approaches to minimize potential side-effects due to the multifunctional roles of TRP channels.
Collapse
|
12
|
Ying Y, Jiang P. Research progress on transient receptor potential melastatin 2 channel in nervous system diseases. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:267-276. [PMID: 34137233 PMCID: PMC8710270 DOI: 10.3724/zdxbyxb-2021-0110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/30/2021] [Indexed: 11/25/2022]
Abstract
Transient receptor potential M2 (TRPM2) ion channel is a non-selective cationic channel that can permeate calcium ions, and plays an important role in neuroinflammation, ischemic reperfusion brain injury, neurodegenerative disease, neuropathic pain, epilepsy and other neurological diseases. In ischemic reperfusion brain injury, TRPM2 mediates neuronal death by modulating the different subunits of glutamate N-methyl-D-aspartic acid receptor in response to calcium/zinc signal. In Alzheimer's disease, TRPM2 is activated by reactive oxygen species generated by β-amyloid peptide to form a malignant positive feedback loop that induces neuronal death and is involved in the pathological process of glial cells by promoting inflammatory response and oxidative stress. In epilepsy, the TRPM2-knockout alleviates epilepsy induced neuronal degeneration by inhibiting autophagy and apoptosis related proteins. The roles of TRPM2 channel in the pathogenesis of various central nervous system diseases and its potential drug development and clinical application prospects are summarized in this review.
Collapse
|
13
|
Zhang H, Zhao S, Yu J, Yang W, Liu Z, Zhang L. Medicinal chemistry perspective of TRPM2 channel inhibitors: where we are and where we might be heading? Drug Discov Today 2020; 25:2326-2334. [PMID: 33065292 DOI: 10.1016/j.drudis.2020.09.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/26/2020] [Accepted: 09/30/2020] [Indexed: 01/06/2023]
Abstract
Transient receptor potential melastatin 2 (TRPM2) is a Ca2+- permeable nonselective cation channel that is involved in diverse biological functions as a cellular sensor for oxidative stress and temperature. It has been considered a promising therapeutic target for the treatment of ischemia/reperfusion (IR) injury, inflammation, cancer, and neurodegenerative diseases. Development of highly potent and selective TRPM2 inhibitors and validation of their use in relevant disease models will advance drug discovery. In this review, we describe the molecular structures and gating mechanism of the TRPM2 channel, and offer a comprehensive review of advances in the discovery of TRPM2 inhibitors. Furthermore, we analyze the properties of reported TRPM2 inhibitors with an emphasis on how specific inhibitors targeting this channel could be better developed.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Siqi Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jie Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wei Yang
- Department of Biophysics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
14
|
Ocak U, Eser Ocak P, Huang L, Xu W, Zuo Y, Li P, Gamdzyk M, Zuo G, Mo J, Zhang G, Zhang JH. Inhibition of mast cell tryptase attenuates neuroinflammation via PAR-2/p38/NFκB pathway following asphyxial cardiac arrest in rats. J Neuroinflammation 2020; 17:144. [PMID: 32366312 PMCID: PMC7199326 DOI: 10.1186/s12974-020-01808-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 04/08/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cardiac arrest survivors suffer from neurological dysfunction including cognitive impairment. Cerebral mast cells, the key regulators of neuroinflammation contribute to neuroinflammation-associated cognitive dysfunction. Mast cell tryptase was demonstrated to have a proinflammatory effect on microglia via the activation of microglial protease-activated receptor-2 (PAR-2). This study investigated the potential anti-neuroinflammatory effect of mast cell tryptase inhibition and the underlying mechanism of PAR-2/p-p38/NFκB signaling following asphyxia-induced cardiac arrest in rats. Methods Adult male Sprague-Dawley rats resuscitated from 10 min of asphyxia-induced cardiac arrest were randomized to four separate experiments including time-course, short-term outcomes, long-term outcomes and mechanism studies. The effect of mast cell tryptase inhibition on asphyxial cardiac arrest outcomes was examined after intranasal administration of selective mast cell tryptase inhibitor (APC366; 50 μg/rat or 150 μg/rat). AC55541 (selective PAR-2 activator; 30 μg/rat) and SB203580 (selective p38 inhibitor; 300 μg/rat) were used for intervention. Short-term neurocognitive functions were evaluated using the neurological deficit score, number of seizures, adhesive tape removal test, and T-maze test, while long-term cognitive functions were evaluated using the Morris water maze test. Hippocampal neuronal degeneration was evaluated by Fluoro-Jade C staining. Results Mast cell tryptase and PAR-2 were dramatically increased in the brain following asphyxia-induced cardiac arrest. The inhibition of mast cell tryptase by APC366 improved both short- and long-term neurological outcomes in resuscitated rats. Such behavioral benefits were associated with reduced expressions of PAR-2, p-p38, NFκB, TNF-α, and IL-6 in the brain as well as less hippocampal neuronal degeneration. The anti-neuroinflammatory effect of APC366 was abolished by AC55541, which when used alone, indeed further exacerbated neuroinflammation, hippocampal neuronal degeneration, and neurologic deficits following cardiac arrest. The deleterious effects aggregated by AC55541 were minimized by p38 inhibitor. Conclusions The inhibition of mast cell tryptase attenuated neuroinflammation, led to less hippocampal neuronal death and improved neurological deficits following cardiac arrest. This effect was at least partly mediated via inhibiting the PAR-2/p-p38/NFκB signaling pathway. Thus, mast cell tryptase might be a novel therapeutic target in the management of neurological impairment following cardiac arrest.
Collapse
Affiliation(s)
- Umut Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Emergency Medicine, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, 16310, Bursa, Turkey.,Department of Emergency Medicine, Bursa City Hospital, 16110, Bursa, Turkey
| | - Pinar Eser Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Uludag University School of Medicine, 16069, Bursa, Turkey
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Weilin Xu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310009, Hangzhou, China
| | - Yuchun Zuo
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Peng Li
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Affiliated Taicang Hospital, Soochow University, Suzhou, Taicang, 215400, Jiangsu, China
| | - Jun Mo
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Guangyu Zhang
- Mass Spectrometry Core Facility, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA. .,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA. .,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA. .,Department of Neurology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
15
|
Cruz-Torres I, Backos DS, Herson PS. Characterization and Optimization of the Novel Transient Receptor Potential Melastatin 2 Antagonist tatM2NX. Mol Pharmacol 2019; 97:102-111. [PMID: 31772034 DOI: 10.1124/mol.119.117549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable channel activated by adenosine diphosphate ribose metabolites and oxidative stress. TRPM2 contributes to neuronal injury in the brain caused by stroke and cardiac arrest among other diseases including pain, inflammation, and cancer. However, the lack of specific inhibitors hinders the study of TRPM2 in brain pathophysiology. Here, we present the design of a novel TRPM2 antagonist, tatM2NX, which prevents ligand binding and TRPM2 activation. We used mutagenesis of tatM2NX to determine the structure-activity relationship and antagonistic mechanism on TRPM2 using whole-cell patch clamp and Calcium imaging in human embryonic kidney 293 cells with stable human TRPM2 expression. We show that tatM2NX inhibits over 90% of TRPM2 channel currents at concentrations as low as 2 μM. Moreover, tatM2NX is a potent antagonist with an IC50 of 396 nM. Our results from tatM2NX mutagenesis indicate that specific residues within the tatM2NX C terminus are required to confer antagonism on TRPM2. Therefore, the peptide tatM2NX represents a new tool for the study of TRPM2 function in cell biology and enhances our understanding of TRPM2 in disease. SIGNIFICANCE STATEMENT: TatM2NX is a potent TRPM2 channel antagonist with the potential for clinical benefit in neurological diseases. This study characterizes interactions of tatM2NX with TRPM2 and the mechanism of action using structure-activity analysis.
Collapse
Affiliation(s)
- I Cruz-Torres
- Departments of Pharmacology (I.C.-T., P.S.H.) and Anesthesiology (P.S.H.) and Neuronal Injury & Plasticity Program (I.C.-T., P.S.H.), University of Colorado School of Medicine, Aurora, Colorado; and Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado (D.S.B.)
| | - D S Backos
- Departments of Pharmacology (I.C.-T., P.S.H.) and Anesthesiology (P.S.H.) and Neuronal Injury & Plasticity Program (I.C.-T., P.S.H.), University of Colorado School of Medicine, Aurora, Colorado; and Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado (D.S.B.)
| | - P S Herson
- Departments of Pharmacology (I.C.-T., P.S.H.) and Anesthesiology (P.S.H.) and Neuronal Injury & Plasticity Program (I.C.-T., P.S.H.), University of Colorado School of Medicine, Aurora, Colorado; and Department of Pharmaceutical Sciences, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado (D.S.B.)
| |
Collapse
|