1
|
Abdel Mageed SS, Rashad AA, Elshaer SS, Elballal MS, Mohammed OA, Darwish SF, Salama RM, Mangoura SA, Al-Noshokaty TM, Gomaa RM, Elesawy AE, El-Demerdash AA, Zaki MB, Abulsoud AI, El-Dakroury WA, Elrebehy MA, Abdel-Reheim MA, Moustafa YM, Gedawy EM, Doghish AS. The emerging role of miRNAs in epilepsy: From molecular signatures to diagnostic potential. Pathol Res Pract 2024; 254:155146. [PMID: 38266457 DOI: 10.1016/j.prp.2024.155146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Epilepsy is a medical condition characterized by intermittent seizures accompanied by changes in consciousness. Epilepsy significantly impairs the daily functioning and overall well-being of affected individuals. Epilepsy is a chronic neurological disorder characterized by recurrent seizures resulting from various dysfunctions in brain activity. The molecular processes underlying changes in neuronal structure, impaired apoptotic responses in neurons, and disruption of regenerative pathways in glial cells in epilepsy remain unknown. MicroRNAs (miRNAs) play a crucial role in regulating apoptosis, autophagy, oxidative stress, neuroinflammation, and the body's regenerative and immune responses. miRNAs have been shown to influence many pathogenic processes in epilepsy including inflammatory responses, neuronal necrosis and apoptosis, dendritic growth, synaptic remodeling, and other processes related to the development of epilepsy. Therefore, the purpose of our current analysis was to determine the role of miRNAs in the etiology and progression of epilepsy. Furthermore, they have been examined for their potential application as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed A Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Shereen Saeid Elshaer
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11823, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Samar F Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Rania M Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt
| | - Safwat Abdelhady Mangoura
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Tohada M Al-Noshokaty
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Rania M Gomaa
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Badr City, P.O. Box 11829, Cairo, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Aya A El-Demerdash
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Yasser M Moustafa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ehab M Gedawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Badr City, P.O. Box 11829, Cairo, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| |
Collapse
|
2
|
Dell'Osso L, Nardi B, Massoni L, Gravina D, Benedetti F, Cremone IM, Carpita B. Neuroprotective Properties of Antiepileptics: What are the Implications for Psychiatric Disorders? Curr Med Chem 2024; 31:3447-3472. [PMID: 37226791 DOI: 10.2174/0929867330666230523155728] [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: 12/31/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 05/26/2023]
Abstract
Since the discovery of the first antiepileptic compound, increasing attention has been paid to antiepileptic drugs (AEDs), and recently, with the understanding of the molecular mechanism underlying cells death, a new interest has revolved around a potential neuroprotective effect of AEDs. While many neurobiological studies in this field have focused on the protection of neurons, growing data are reporting how exposure to AEDs can also affect glial cells and the plastic response underlying recovery; however, demonstrating the neuroprotective abilities of AEDs remains a changeling task. The present work aims to summarize and review the literature available on the neuroprotective properties of the most commonly used AEDs. Results highlighted how further studies should investigate the link between AEDs and neuroprotective properties; while many studies are available on valproate, results for other AEDs are very limited and the majority of the research has been carried out on animal models. Moreover, a better understanding of the biological basis underlying neuro-regenerative defects may pave the way for the investigation of further therapeutic targets and eventually lead to an improvement in the actual treatment strategies.
Collapse
Affiliation(s)
- Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Benedetta Nardi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Leonardo Massoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Davide Gravina
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Francesca Benedetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Ivan Mirko Cremone
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Barbara Carpita
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| |
Collapse
|
3
|
Jia D, Wang F, Bai Z, Chen X. BDNF-TrkB/proBDNF-p75 NTR pathway regulation by lipid emulsion rescues bupivacaine-induced central neurotoxicity in rats. Sci Rep 2023; 13:18364. [PMID: 37884604 PMCID: PMC10603093 DOI: 10.1038/s41598-023-45572-8] [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: 04/10/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023] Open
Abstract
Bupivacaine (BPV) can cause severe central nervous system toxicity when absorbed into the blood circulation system. Rapid intravenous administration of lipid emulsion (LE) could be used to treat local anaesthetic toxicity. This study aimed to investigate the mechanism by which the BDNF-TrkB/proBDNF-p75NTR pathway regulation by LE rescues BPV induced neurotoxicity in hippocampal neurons in rats. Seven- to nine-day-old primary cultured hippocampal neurons were randomly divided into 6 groups: the blank control group (Ctrl), the bupivacaine group (BPV), the lipid emulsion group (LE), the bupivacaine + lipid emulsion group (BPV + LE), the bupivacaine + lipid emulsion + tyrosine kinase receptor B (TrkB) inhibitor group (BPV + LE + K252a), the bupivacaine + lipid emulsion + p75 neurotrophic factor receptor (p75NTR) inhibitor group (BPV + LE + TAT-Pep5). All hippocampal neurons were incubated for 24 h, and their growth state was observed by light microscopy. The relative TrkB and p75NTR mRNA levels were detected by real-time PCR. The protein expression levels of brain-derived neurotrophic factor (BDNF), proBDNF, TrkB, p75NTR and cleaved caspase-3 were detected by western blotting. The results showed that primary hippocampal neuron activity was reduced by BPV. As administration of LE elevated hippocampal neuronal activity, morphology was also somewhat improved. The protein expression and mRNA levels of TrkB and p75NTR were decreased when BPV induced hippocampal neuronal toxicity, while the expression of BDNF was increased. At the same time, BPV increased the original generation of cleaved caspase-3 protein content by hippocampal neurons, while the content of cleaved caspase-3 protein in hippocampal neurons cotreated with LE and BPV was decreased. Thus, this study has revealed LE may reduce apoptosis and promote survival of hippocampal neurons by regulating the BDNF-TrkB pathway and the proBDNF-p75NTR pathway to rescue BPV induced central neurotoxicity in rats.
Collapse
Affiliation(s)
- Danting Jia
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Fang Wang
- Department of Anaesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, Ningxia, China
| | - Zhixia Bai
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xuexin Chen
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
4
|
Arab HH, Eid AH, Yahia R, Alsufyani SE, Ashour AM, El-Sheikh AAK, Darwish HW, Saad MA, Al-Shorbagy MY, Masoud MA. Targeting Autophagy, Apoptosis, and SIRT1/Nrf2 Axis with Topiramate Underlies Its Neuroprotective Effect against Cadmium-Evoked Cognitive Deficits in Rats. Pharmaceuticals (Basel) 2023; 16:1214. [PMID: 37765022 PMCID: PMC10535870 DOI: 10.3390/ph16091214] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 09/29/2023] Open
Abstract
Cadmium is an environmental toxicant that instigates cognitive deficits with excessive glutamate excitatory neuroactivity in the brain. Topiramate, a glutamate receptor antagonist, has displayed favorable neuroprotection against epilepsy, cerebral ischemia, and Huntington's disease; however, its effect on cadmium neurotoxicity remains to be investigated. In this study, topiramate was tested for its potential to combat the cognitive deficits induced by cadmium in rats with an emphasis on hippocampal oxidative insult, apoptosis, and autophagy. After topiramate intake (50 mg/kg/day; p.o.) for 8 weeks, behavioral disturbances and molecular changes in the hippocampal area were explored. Herein, Morris water maze, Y-maze, and novel object recognition test revealed that topiramate rescued cadmium-induced memory/learning deficits. Moreover, topiramate significantly lowered hippocampal histopathological damage scores. Mechanistically, topiramate significantly replenished hippocampal GLP-1 and dampened Aβ42 and p-tau neurotoxic cues. Notably, it significantly diminished hippocampal glutamate content and enhanced acetylcholine and GABA neurotransmitters. The behavioral recovery was prompted by hippocampal suppression of the pro-oxidant events with notable activation of SIRT1/Nrf2/HO-1 axis. Moreover, topiramate inactivated GSK-3β and dampened the hippocampal apoptotic changes. In tandem, stimulation of hippocampal pro-autophagy events, including Beclin 1 upregulation, was triggered by topiramate that also activated AMPK/mTOR pathway. Together, the pro-autophagic, antioxidant, and anti-apoptotic features of topiramate contributed to its neuroprotective properties in rats intoxicated with cadmium. Therefore, it may be useful to mitigate cadmium-induced cognitive deficits.
Collapse
Affiliation(s)
- Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (H.H.A.); (S.E.A.)
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Ahmed H. Eid
- Department of Pharmacology, Egyptian Drug Authority (EDA)—Formerly NODCAR, Giza 12654, Egypt; (A.H.E.); (R.Y.); (M.A.M.)
| | - Rania Yahia
- Department of Pharmacology, Egyptian Drug Authority (EDA)—Formerly NODCAR, Giza 12654, Egypt; (A.H.E.); (R.Y.); (M.A.M.)
| | - Shuruq E. Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (H.H.A.); (S.E.A.)
| | - Ahmed M. Ashour
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al Qura University, P.O. Box 13578, Makkah 21955, Saudi Arabia;
| | - Azza A. K. El-Sheikh
- Basic Health Sciences Department, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Hany W. Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 11451, Riyadh 11451, Saudi Arabia;
| | - Muhammed A. Saad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Muhammad Y. Al-Shorbagy
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Marwa A. Masoud
- Department of Pharmacology, Egyptian Drug Authority (EDA)—Formerly NODCAR, Giza 12654, Egypt; (A.H.E.); (R.Y.); (M.A.M.)
| |
Collapse
|
5
|
Ozkula S, Jafarova Demirkapu M, Yananli HR, Aydin B, Nacar C, Cabadak H. The effect of acute topiramate administration on morphine withdrawal syndrome and brain-derived neurotrophic factor in central nervous system. Neurol Res 2023:1-8. [PMID: 37105528 DOI: 10.1080/01616412.2023.2203611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
OBJECTIVES Nucleus accumbens plays an important role in opioid addiction. Topiramate, increases postsynaptic gamma-aminobutyric acid receptor activity and antagonizes glutamatergic activity. Brain-derived neurotrophic factor (BDNF), which plays a key role in synaptic plasticity, is produced from proBDNF. The aim of this study is to investigate the effects of 100 µM topiramate applied into the lateral ventricle or nucleus accumbens on naloxone-induced morphine withdrawal and the BDNF/proBDNF ratio in the frontal cortex. METHODS In the study, 36 adult male Wistar rats weighing 250-350 g were used. Morphine dependence was created with morphine pellets following guide cannula implantations. Withdrawal findings were evaluated in naloxone-induced morphine withdrawal syndrome following topiramate administration, and locomotor activity measurements were performed simultaneously. The brains of sacrificed animals were removed for determination of BDNF/proBDNF ratio. RESULTS Topiramate administered by either route significantly suppressed the number of jumps in morphine withdrawal. Topiramate applied into the nucleus accumbens significantly reduced stereotypical behavior in morphine withdrawal, but did not cause any changes in other locomotor activity behaviors. Topiramate applied into the lateral ventricle significantly decreased the BDNF/proBDNF ratio, whereas administered into the nucleus accumbens significantly increased this ratio. CONCLUSION The findings of this study indicate that topiramate administered into the lateral ventricle and nucleus accumbens reduces naloxone-induced morphine withdrawal symptoms, stereotypical locomotor activity, and changes the BDNF/proBDNF ratio.
Collapse
Affiliation(s)
- Songul Ozkula
- Department of Pharmacology, Suleyman Demirel University Faculty of Medicine, Isparta, Turkey
| | | | - Hasan Raci Yananli
- Department of Pharmacology, Marmara University School of Medicine, Istanbul, Turkey
| | - Banu Aydin
- Department of Biophysics, Marmara University School of Medicine, Istanbul, Turkey
| | - Cevdet Nacar
- Department of Biophysics, Marmara University School of Medicine, Istanbul, Turkey
| | - Hulya Cabadak
- Department of Biophysics, Marmara University School of Medicine, Istanbul, Turkey
| |
Collapse
|
6
|
Lu M, He X, Jiao Z, Hu Z, Guo Z, Dai S, Wang H, Xu D. The upregulation of glutamate decarboxylase 67 against hippocampal excitability damage in male fetal rats by prenatal caffeine exposure. ENVIRONMENTAL TOXICOLOGY 2022; 37:2703-2717. [PMID: 35917217 DOI: 10.1002/tox.23630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/25/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
As a kind of xanthine alkaloid, caffeine is widely present in beverages, food, and analgesic drugs. Our previous studies have shown that prenatal caffeine exposure (PCE) can induce programmed hypersensitivity of the hypothalamic-pituitary-adrenal (HPA) axis in offspring rats, which is involved in developing many chronic adult diseases. The present study further examined the potential molecular mechanism and toxicity targets of hippocampal dysfunction, which might mediate the programmed hypersensitivity of the HPA axis in offspring. Pregnant rats were intragastrically administered with 0, 30, and 120 mg/kg/day caffeine from gestational days (GD) 9-20, and the fetal rats were extracted at GD20. Rat fetal hippocampal H19-7/IGF1R cell line was treated with caffeine, adenosine A2A receptor (A2AR) agonist (CGS-21680) or adenylate cyclase agonist (forskolin) plus caffeine. Compared with the control group, hippocampal neurons of male fetal rats by PCE displayed increased apoptosis and reduced synaptic plasticity, whereas glutamate decarboxylase 67 (GAD67) expression was increased. Moreover, the expression of A2AR was down-regulated, PCE inhibited the cAMP/PKA/CREB/BDNF/TrkB pathway. Furthermore, the results in vitro were consistent with the in vivo study. Both CGS21680 and forskolin could reverse the above alteration caused by caffeine. These results indicated that PCE inhibits the BDNF pathway and mediates the hippocampus's glutamate (Glu) excitotoxicity. The compensatory up-regulation of GAD67 unbalanced the Glu/gamma-aminobutyric acid (GABA)ergic output, leading to the impaired negative feedback to the hypothalamus and hypersensitivity of the HPA axis.
Collapse
Affiliation(s)
- Mengxi Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xia He
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zhexiao Jiao
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zewen Hu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zijing Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Shiyun Dai
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Dan Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
7
|
Dihydromyricetin ameliorates social isolation-induced anxiety by modulating mitochondrial function, antioxidant enzymes, and BDNF. Neurobiol Stress 2022; 21:100499. [DOI: 10.1016/j.ynstr.2022.100499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/08/2022] Open
|
8
|
Cristiano C, Cuozzo M, Coretti L, Liguori F, Cimmino F, Turco L, Avagliano C, Aviello G, Mollica M, Lembo F, Russo R. Oral sodium butyrate supplementation ameliorates paclitaxel-induced behavioral and intestinal dysfunction. Biomed Pharmacother 2022; 153:113528. [DOI: 10.1016/j.biopha.2022.113528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
|
9
|
Lemon N, Canepa E, Ilies MA, Fossati S. Carbonic Anhydrases as Potential Targets Against Neurovascular Unit Dysfunction in Alzheimer’s Disease and Stroke. Front Aging Neurosci 2021; 13:772278. [PMID: 34867298 PMCID: PMC8635164 DOI: 10.3389/fnagi.2021.772278] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/20/2021] [Indexed: 12/23/2022] Open
Abstract
The Neurovascular Unit (NVU) is an important multicellular structure of the central nervous system (CNS), which participates in the regulation of cerebral blood flow (CBF), delivery of oxygen and nutrients, immunological surveillance, clearance, barrier functions, and CNS homeostasis. Stroke and Alzheimer Disease (AD) are two pathologies with extensive NVU dysfunction. The cell types of the NVU change in both structure and function following an ischemic insult and during the development of AD pathology. Stroke and AD share common risk factors such as cardiovascular disease, and also share similarities at a molecular level. In both diseases, disruption of metabolic support, mitochondrial dysfunction, increase in oxidative stress, release of inflammatory signaling molecules, and blood brain barrier disruption result in NVU dysfunction, leading to cell death and neurodegeneration. Improved therapeutic strategies for both AD and stroke are needed. Carbonic anhydrases (CAs) are well-known targets for other diseases and are being recently investigated for their function in the development of cerebrovascular pathology. CAs catalyze the hydration of CO2 to produce bicarbonate and a proton. This reaction is important for pH homeostasis, overturn of cerebrospinal fluid, regulation of CBF, and other physiological functions. Humans express 15 CA isoforms with different distribution patterns. Recent studies provide evidence that CA inhibition is protective to NVU cells in vitro and in vivo, in models of stroke and AD pathology. CA inhibitors are FDA-approved for treatment of glaucoma, high-altitude sickness, and other indications. Most FDA-approved CA inhibitors are pan-CA inhibitors; however, specific CA isoforms are likely to modulate the NVU function. This review will summarize the literature regarding the use of pan-CA and specific CA inhibitors along with genetic manipulation of specific CA isoforms in stroke and AD models, to bring light into the functions of CAs in the NVU. Although pan-CA inhibitors are protective and safe, we hypothesize that targeting specific CA isoforms will increase the efficacy of CA inhibition and reduce side effects. More studies to further determine specific CA isoforms functions and changes in disease states are essential to the development of novel therapies for cerebrovascular pathology, occurring in both stroke and AD.
Collapse
Affiliation(s)
- Nicole Lemon
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Elisa Canepa
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Marc A. Ilies
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Temple University, Philadelphia, PA, United States
| | - Silvia Fossati
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- *Correspondence: Silvia Fossati,
| |
Collapse
|
10
|
Sarkar P, Jayaraj P, Patwardhan K, Yeole S, Das S, Somaiya Y, Desikan R, Thirumurugan K. In Silico Analysis to Link Insulin Resistance, Obesity and Ageing with Alzheimer's Disease. J Mol Neurosci 2021; 71:2608-2617. [PMID: 34227035 DOI: 10.1007/s12031-021-01875-x] [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: 12/16/2020] [Accepted: 06/16/2021] [Indexed: 01/09/2023]
Abstract
The process of ageing accompanies several metabolic diseases. With ageing, fats accumulate to increase the visceral and abdominal adiposity leading to hyperinsulinemia, insulin resistance, obesity and several other diseases. Drosophila melanogaster is often used to study the ageing process and its related disorders. Therefore, in this study, we performed an in silico analysis to relate the process of ageing and insulin resistance. We analysed the data of insulin-resistant Drosophila from the GEO database and compared it with the data from the literature survey. We observed that 98 genes were common in both the models, and they showed gene modulations related to metabolic pathways, fatty acid metabolism, insulin resistance and neural receptor-ligand binding pathways. Analysis of the REACTOME database against human data revealed that the TRKB signalling pathway is commonly affected. The TRKB-mediated BDNF pathway is a major regulator of memory loss. We further analysed the common genes in Alzheimer's disease and compared the fly data with human data to identify the diseases related to these common genes. Then, we performed a literature survey to provide protective mechanisms for the TRKB signalling pathway activation, mediated through polyphenols. We treated the flies with sesamol-conjugated lipoic acid derivative (a phenolic compound) at hormetic doses to evaluate its effect on the memory of flies.
Collapse
Affiliation(s)
- Priyanka Sarkar
- Structural Biology Lab, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Premkumar Jayaraj
- Technology Tower, Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, India
| | - Ketaki Patwardhan
- Structural Biology Lab, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Samiksha Yeole
- Structural Biology Lab, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Sourajit Das
- Structural Biology Lab, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Yash Somaiya
- Structural Biology Lab, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Rajagopal Desikan
- Technology Tower, Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, India
| | - Kavitha Thirumurugan
- Structural Biology Lab, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
11
|
Soleimani Meigoni Z, Jabari F, Motaghinejad M, Motevalian M. Protective effects of forced exercise against topiramate-induced cognition impairment and enhancement of its antiepileptic activity: molecular and behavioral evidences. Int J Neurosci 2021; 132:1198-1209. [PMID: 33428483 DOI: 10.1080/00207454.2021.1873979] [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] [Indexed: 02/02/2023]
Abstract
Propose/aim of study: Forced exercise can act as a neuroprotective factor and cognitive enhancer. The aim of the current study was to evaluate the effects of forced exercise on topiramate (TPM) induced cognitive impairment and also on TPM anti-seizure activity and neurodegeneration status after seizure.Material and method: Forty adult male rats were divided into four groups receiving normal saline, TPM (100 mg/kg), TPM in combination with forced exercise and forced exercise only respectively for 21 days. MWM test, and PTZ induced seizure were used and some oxidative, inflammatory and apoptotic biomarkers were measured for assessment of experimental animals.Results: Forced exercise in combination with TPM could abolish the TPM induced cognitive impairment and potentiates its anti-seizure activity. Also forced exercise in combination with TPM decreased malondialdehyde (MDA), tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) and Bax protein, while caused increase in superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione reductase (GR) activities after PTZ administration.Conclusion: It seems that forced exercise could act as an adjunct therapy with TPM for management of induced cognitive impairment and can also potentiate TPM antiepileptic and neuroprotective effects.
Collapse
Affiliation(s)
- Zahra Soleimani Meigoni
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Jabari
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Manijeh Motevalian
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Chen S, Zou Q, Guo Q, Chen Y, Kuang X, Zhang Y, Liu Y, Wu W, Li G, Tu L, Tong J, Li S, Ma L, Li Q. SPARC Knockdown Reduces Glutamate-Induced HT22 Hippocampal Nerve Cell Damage by Regulating Autophagy. Front Neurosci 2021; 14:581441. [PMID: 33584170 PMCID: PMC7874057 DOI: 10.3389/fnins.2020.581441] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein involved in the extracellular matrix and interactions between cells during neural development of the central nervous system (CNS). Oxidative glutamate toxicity is involved in CNS diseases, including epilepsy, Alzheimer’s disease, and ischemic stroke. However, the molecular mechanism of nerve injury is not fully understood in CNS diseases. Herein, the glutamate-induced nerve damage model was used to explore the molecular mechanisms affecting nerve damage. The levels of SPARC and autophagy were increased in glutamate-induced HT22 hippocampal nerve injury. In summary, the current study confirmed that SPARC regulates autophagy in HT22 hippocampal nerve cells, and its knockdown reduces the glutamate-induced HT22 hippocampal nerve injury by inhibiting autophagy. These findings suggested that SPARC plays a crucial role in nerve injury of CNS diseases.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Qin Zou
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qiang Guo
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Yongmin Chen
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Xi Kuang
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Hainan Health Vocational College, Haikou, China
| | - Yukang Zhang
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Yan Liu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Wengang Wu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Ge Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Linzhi Tu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Jingyi Tong
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Songrong Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Lin Ma
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Qifu Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| |
Collapse
|
13
|
Baek SY, Kim MR. Neuroprotective Effect of Carotenoid-Rich Enteromorpha prolifera Extract via TrkB/Akt Pathway against Oxidative Stress in Hippocampal Neuronal Cells. Mar Drugs 2020; 18:md18070372. [PMID: 32707633 PMCID: PMC7404284 DOI: 10.3390/md18070372] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
In this study, we found that E. prolifera extract (EAEP) exhibits neuroprotective effects in oxidative stress-induced neuronal cells. EAEP improved cell viability as well as attenuated the formation of intracellular reactive oxygen species (ROS) and apoptotic bodies in glutamate-treated hippocampal neuronal cells (HT-22). Furthermore, EAEP improved the expression of brain-derived neurotrophic factor (BDNF) and antioxidant enzymes such as heme oxygenase-1 (HO-1), NAD(P)H quinine oxidoreductase-1 (NQO-1), and glutamate-cysteine ligase catalytic subunit (GCLC) via the tropomyosin-related kinase receptor B/ protein kinase B (TrkB/Akt) signaling pathway. In contrast, the pre-incubation of K252a, a TrkB inhibitor, or MK-2206, an Akt-selective inhibitor, ameliorated the neuroprotective effects of EAEP in oxidative stress-induced neuronal cells. These results suggest that EAEP protects neuronal cells against oxidative stress-induced apoptosis by upregulating the expression of BDNF and antioxidant enzymes via the activation of the TrkB/Akt pathway. In conclusion, such an effect of EAEP, which is rich in carotenoid-derived compounds, may justify its application as a food supplement in the prevention and treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Mee Ree Kim
- Correspondence: ; Tel.: +82-42-821-6837; Fax: +82-42-821-8887
| |
Collapse
|
14
|
Hannan MA, Dash R, Sohag AAM, Haque MN, Moon IS. Neuroprotection Against Oxidative Stress: Phytochemicals Targeting TrkB Signaling and the Nrf2-ARE Antioxidant System. Front Mol Neurosci 2020; 13:116. [PMID: 32714148 PMCID: PMC7346762 DOI: 10.3389/fnmol.2020.00116] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) plays a critical role in the pathophysiology of several brain-related disorders, including neurodegenerative diseases and ischemic stroke, which are the major causes of dementia. The Nrf2-ARE (nuclear factor erythroid 2-related factor 2/antioxidant responsive element antioxidant) system, the primary cellular defense against OS, plays an essential role in neuroprotection by regulating the expressions of antioxidant molecules and enzymes. However, simultaneous events resulting in the overproduction of reactive oxygen species (ROS) and deregulation of the Nrf2-ARE system damage essential cell components and cause loss of neuron structural and functional integrity. On the other hand, TrkB (tropomyosin-related kinase B) signaling, a classical neurotrophin signaling pathway, regulates neuronal survival and synaptic plasticity, which play pivotal roles in memory and cognition. Also, TrkB signaling, specifically the TrkB/PI3K/Akt (TrkB/phosphatidylinositol 3 kinase/protein kinase B) pathway promotes the activation and nuclear translocation of Nrf2, and thus, confers neuroprotection against OS. However, the TrkB signaling pathway is also known to be downregulated in brain disorders due to lack of neurotrophin support. Therefore, activations of TrkB and the Nrf2-ARE signaling system offer a potential approach to the design of novel therapeutic agents for brain disorders. Here, we briefly overview the development of OS and the association between OS and the pathogenesis of neurodegenerative diseases and brain injury. We propose the cellular antioxidant defense and TrkB signaling-mediated cell survival systems be considered pharmacological targets for the treatment of neurodegenerative diseases, and review the literature on the neuroprotective effects of phytochemicals that can co-activate these neuronal defense systems.
Collapse
Affiliation(s)
- Md. Abdul Hannan
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
| | - Abdullah Al Mamun Sohag
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Nazmul Haque
- Department of Fisheries Biology and Genetics, Patuakhali Science and Technology University, Patuakhali, Bangladesh
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
| |
Collapse
|
15
|
Wang W, Yang L, Liu T, Ma Y, Huang S, He M, Wang J, Wen A, Ding Y. Corilagin ameliorates sleep deprivation-induced memory impairments by inhibiting NOX2 and activating Nrf2. Brain Res Bull 2020; 160:141-149. [DOI: 10.1016/j.brainresbull.2020.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/07/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
|
16
|
Bonnet U, Wiemann M. Topiramate Decelerates Bicarbonate-Driven Acid-Elimination of Human Neocortical Neurons: Strategic Significance for its Antiepileptic, Antimigraine and Neuroprotective Properties. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:264-275. [PMID: 32496992 DOI: 10.2174/1871527319666200604173208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Mammalian central neurons regulate their intracellular pH (pHi) strongly and even slight pHi-fluctuations can influence inter-/intracellular signaling, synaptic plasticity and excitability. OBJECTIVE For the first time, we investigated topiramate´s (TPM) influence on pHi-behavior of human central neurons representing a promising target for anticonvulsants and antimigraine drugs. METHODS In slice-preparations of tissue resected from the middle temporal gyrus of five adults with intractable temporal lobe epilepsy, BCECF-AM-loaded neocortical pyramidal-cells were investigated by fluorometry. The pHi-regulation was estimated by using the recovery-slope from intracellular acidification after an Ammonium-Prepulse (APP). RESULTS Among 17 pyramidal neurons exposed to 50 μM TPM, seven (41.24%) responded with an altered resting-pHi (7.02±0.12), i.e., acidification of 0.01-0.03 pH-units. The more alkaline the neurons, the greater the TPM-related acidifications (r=0.7, p=0.001, n=17). The recovery from APPacidification was significantly slowed under TPM (p<0.001, n=5). Further experiments using nominal bicarbonate-free (n=2) and chloride-free (n=2) conditions pointed to a modulation of the HCO3 -- driven pHi-regulation by TPM, favoring a stimulation of the passive Cl-/HCO3 --antiporter (CBT) - an acid-loader predominantly in more alkaline neurons. CONCLUSION TPM modulated the bicarbonate-driven pHi-regulation, just as previously described in adult guinea-pig hippocampal neurons. We discussed the significance of the resulting subtle acidifications for beneficial antiepileptic, antimigraine and neuroprotective effects as well as for unwanted cognitive deficits.
Collapse
Affiliation(s)
- Udo Bonnet
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Evangelisches Krankenhaus Castrop-Rauxel, Academic Teaching Hospital of the University Duisburg-Essen, Castrop-Rauxel, Germany.,Department of Psychiatry and Psychotherapy, Faculty of Medicine, LVR-Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martin Wiemann
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany.,IBE R&D gGmbH, Institute for Lung Health, D-48149 Munster, Germany
| |
Collapse
|
17
|
Marafi D, Mitani T, Isikay S, Hertecant J, Almannai M, Manickam K, Abou Jamra R, El-Hattab AW, Rajah J, Fatih JM, Du H, Karaca E, Bayram Y, Punetha J, Rosenfeld JA, Jhangiani SN, Boerwinkle E, Akdemir ZC, Erdin S, Hunter JV, Gibbs RA, Pehlivan D, Posey JE, Lupski JR. Biallelic GRM7 variants cause epilepsy, microcephaly, and cerebral atrophy. Ann Clin Transl Neurol 2020; 7:610-627. [PMID: 32286009 PMCID: PMC7261753 DOI: 10.1002/acn3.51003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 01/29/2023] Open
Abstract
Objective Defects in ion channels and neurotransmitter receptors are implicated in developmental and epileptic encephalopathy (DEE). Metabotropic glutamate receptor 7 (mGluR7), encoded by GRM7, is a presynaptic G‐protein‐coupled glutamate receptor critical for synaptic transmission. We previously proposed GRM7 as a candidate disease gene in two families with neurodevelopmental disorders (NDDs). One additional family has been published since. Here, we describe three additional families with GRM7 biallelic variants and deeply characterize the associated clinical neurological and electrophysiological phenotype and molecular data in 11 affected individuals from six unrelated families. Methods Exome sequencing and family‐based rare variant analyses on a cohort of 220 consanguineous families with NDDs revealed three families with GRM7 biallelic variants; three additional families were identified through literature search and collaboration with a clinical molecular laboratory. Results We compared the observed clinical features and variants of 11 affected individuals from the six unrelated families. Identified novel deleterious variants included two homozygous missense variants (c.2671G>A:p.Glu891Lys and c.1973G>A:p.Arg685Gln) and one homozygous stop‐gain variant (c.1975C>T:p.Arg659Ter). Developmental delay, neonatal‐ or infantile‐onset epilepsy, and microcephaly were universal. Three individuals had hypothalamic–pituitary–axis dysfunction without pituitary structural abnormality. Neuroimaging showed cerebral atrophy and hypomyelination in a majority of cases. Two siblings demonstrated progressive loss of myelination by 2 years in both and an acquired microcephaly pattern in one. Five individuals died in early or late childhood. Conclusion Detailed clinical characterization of 11 individuals from six unrelated families demonstrates that rare biallelic GRM7 pathogenic variants can cause DEEs, microcephaly, hypomyelination, and cerebral atrophy.
Collapse
Affiliation(s)
- Dana Marafi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Department of Pediatrics, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait
| | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Sedat Isikay
- Department of Physiotherapy and Rehabilitation, School of Health Sciences, Hasan Kalyoncu University, Gaziantep, 27000, Turkey
| | - Jozef Hertecant
- Pediatric Metabolic and Genetics Division, Tawam Hospital, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mohammed Almannai
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Kandamurugu Manickam
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Rami Abou Jamra
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Ayman W El-Hattab
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Jaishen Rajah
- Sheikh Khalifa Medical City (SKMC), P.O. Box: 51900, Abu Dhabi, United Arab Emirates
| | - Jawid M Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Haowei Du
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Ender Karaca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Yavuz Bayram
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Jaya Punetha
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030.,Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas
| | - Zeynep C Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Serkan Erdin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jill V Hunter
- Texas Children's Hospital, Houston, Texas, 77030.,Department of Radiology, Baylor College of Medicine, Houston, Texas, 77030
| | - Richard A Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Texas Children's Hospital, Houston, Texas, 77030.,Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030.,Texas Children's Hospital, Houston, Texas, 77030.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030
| |
Collapse
|
18
|
Yan T, Sun Y, Wu B, Xiao F, Bi K, Sun B, Jia Y. Red wine polyphenols reverse depressive-like behaviors in mice induced by repeated corticosterone treatment. CIÊNCIA E TÉCNICA VITIVINÍCOLA 2019. [DOI: 10.1051/ctv/201934020115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The aim of this study was to investigate the antidepressant-like effect of red wine phenolic extracts in mouse model exposed to exogenous corticosterone. The results showed that 3-week corticosterone injections caused depression-like behavior in mice, as indicated by the significant decrease in sucrose consumption and increase immobility time in the forced swimming test (FST). Red wine phenolic extracts treatment significantly reduced serum corticosterone levels. Moreover, it was found that red wine phenolic extract increased the brain-derived neurotrophic factor protein (BNDF) and tropomyosin-related kinase B (TrkB) phosphorylation and cAMP-responsive element binding protein (CREB) phosphorylation levels in the hippocampus and prefrontal cortex. However, K252a, an inhibitor of TrkB, completely abolished those antidepressant-like effects. These results suggested that the red wine phenolic extracts produce an antidepressant-like effect in corticosteronetreated mice, at least in part, which is possibly mediated by modulating hypothalamic-pituitary-adrenal (HPA) axis, BDNF, TrkB and CREB phosphorylation levels in the brain region of mice.
Collapse
|
19
|
Yan T, Sun Y, Xiao F, Wu B, Bi K, He B, Jia Y. Schisandrae Chinensis Fructus inhibits behavioral deficits induced by sleep deprivation and chronic unpredictable mild stress via increased signaling of brain‐derived neurotrophic factor. Phytother Res 2019; 33:3177-3190. [DOI: 10.1002/ptr.6489] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/04/2019] [Accepted: 08/12/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Tingxu Yan
- School of Functional Food and WineShenyang Pharmaceutical University Shenyang China
| | - Yingying Sun
- School of Traditional Chinese Materia MedicaShenyang Pharmaceutical University Shenyang China
| | - Feng Xiao
- School of Functional Food and WineShenyang Pharmaceutical University Shenyang China
| | - Bo Wu
- School of Functional Food and WineShenyang Pharmaceutical University Shenyang China
| | - Kaishun Bi
- School of PharmacyShenyang Pharmaceutical University Shenyang China
| | - Bosai He
- School of Functional Food and WineShenyang Pharmaceutical University Shenyang China
| | - Ying Jia
- School of Functional Food and WineShenyang Pharmaceutical University Shenyang China
| |
Collapse
|
20
|
Zhao Y, Wang J, Du J, Li B, Gou X, Liu J, Hou L, Sang H, Deng B. TAT-Ngn2 Enhances Cognitive Function Recovery and Regulates Caspase-Dependent and Mitochondrial Apoptotic Pathways After Experimental Stroke. Front Cell Neurosci 2018; 12:475. [PMID: 30618628 PMCID: PMC6302814 DOI: 10.3389/fncel.2018.00475] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022] Open
Abstract
Neurogenin-2 (Ngn2) is a basic helix-loop-helix (bHLH) transcription factor that contributes to the identification and specification of neuronal fate during neurogenesis. In our previous study, we found that Ngn2 plays an important role in alleviating neuronal apoptosis, which may be viewed as an attractive candidate target for the treatment of cerebral ischemia. However, novel strategies require an understanding of the function and mechanism of Ngn2 in mature hippocampal neurons after global cerebral ischemic injury. Here, we found that the expression of Ngn2 decreased in the hippocampus after global cerebral ischemic injury in mice and in primary hippocampal neurons after oxygen glucose deprivation (OGD) injury. Then, transactivator of transcription (TAT)-Ngn2, which was constructed by fusing a TAT domain to Ngn2, was effectively transported and incorporated into hippocampal neurons after intraperitoneal (i.p.) injection and enhanced cognitive functional recovery in the acute stage after reperfusion. Furthermore, TAT-Ngn2 alleviated hippocampal neuronal damage and apoptosis, and inhibited the cytochrome C (CytC) leak from the mitochondria to the cytoplasm through regulating the expression levels of brain-derived neurotrophic factor (BDNF), phosphorylation tropomyosin-related kinase B (pTrkB), Bcl-2, Bax and cleaved caspase-3 after reperfusion injury in vivo and in vitro. These findings suggest that the downregulation of Ngn2 expression may have an important role in triggering brain injury after ischemic stroke and that the neuroprotection of TAT-Ngn2 against stroke might involve the modulation of BDNF-TrkB signaling that regulates caspase-dependent and mitochondrial apoptotic pathways, which may be an attractive therapeutic strategy for cerebral ischemic injury.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Hygienic Toxicology, Public Health College, Harbin Medical University, Harbin, China.,Department of Anesthesiology, Heilongjiang Provincial Hospital, Harbin, China
| | - Jinling Wang
- Department of Emergency, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Jiwei Du
- Department of Nursing, Xiang'an Hospital, Xiamen University, Xiamen, China
| | - Baixiang Li
- Department of Hygienic Toxicology, Public Health College, Harbin Medical University, Harbin, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Jiannan Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Lichao Hou
- Department of Anesthesiology, Xiang'an Hospital, Xiamen University, Xiamen, China
| | - Hanfei Sang
- Department of Anesthesiology, Xiang'an Hospital, Xiamen University, Xiamen, China
| | - Bin Deng
- Department of Anesthesiology, Xiang'an Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
21
|
Chen SF, Jou SB, Chen NC, Chuang HY, Huang CR, Tsai MH, Tan TY, Tsai WC, Chang CC, Chuang YC. Serum Levels of Brain-Derived Neurotrophic Factor and Insulin-Like Growth Factor 1 Are Associated With Autonomic Dysfunction and Impaired Cerebral Autoregulation in Patients With Epilepsy. Front Neurol 2018; 9:969. [PMID: 30524358 PMCID: PMC6256185 DOI: 10.3389/fneur.2018.00969] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Brain-derived neurotrophic factor (BDNF) and insulin-like growth factor 1 (IGF-1) may regulate the autonomic nervous system (ANS) in epilepsy. The present study investigated the role of IGF-1 and BDNF in the regulation of autonomic functions and cerebral autoregulation in patients with epilepsy. Methods: A total of 57 patients with focal epilepsy and 35 healthy controls were evaluated and their sudomotor, cardiovagal, and adrenergic functions were assessed using a battery of ANS function tests, including the deep breathing, Valsalva maneuver, head-up tilting, and Q-sweat tests. Cerebral autoregulation was measured by transcranial doppler during the breath-holding test and the Valsalva maneuver. Interictal serum levels of BDNF and IGF-1 were measured with enzyme-linked immunosorbent assay kits. Results: During interictal period, reduced serum levels of BDNF and IGF-1, impaired autonomic functions, and decreased cerebral autoregulation were noted in patients with epilepsy compared with healthy controls. Reduced serum levels of BDNF correlated with age, adrenergic and sudomotor function, overall autonomic dysfunction, and the autoregulation index calculated in Phase II of the Valsalva maneuver, and showed associations with focal to bilateral tonic-clonic seizures. Reduced serum levels of IGF-1 were found to correlate with age and cardiovagal function, a parameter of cerebral autoregulation (the breath-hold index). Patients with a longer history of epilepsy, higher seizure frequency, and temporal lobe epilepsy had lower serum levels of IGF-1. Conclusions: Long-term epilepsy and severe epilepsy, particularly temporal lobe epilepsy, may perturb BDNF and IGF-1 signaling in the central autonomic system, contributing to the autonomic dysfunction and impaired cerebral autoregulation observed in patients with focal epilepsy.
Collapse
Affiliation(s)
- Shu-Fang Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shuo-Bin Jou
- Department of Neurology, Mackay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Nai-Ching Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-Yi Chuang
- Department of Occupational and Environmental Medicine, Kaohsiung Medical University Hospital and School of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Ren Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Meng-Han Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Teng-Yeow Tan
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wan-Chen Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chiung-Chih Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yao-Chung Chuang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
22
|
Das D, Biswal S, Barhwal KK, Chaurasia OP, Hota SK. Kaempferol Inhibits Extra-synaptic NMDAR-Mediated Downregulation of TRkβ in Rat Hippocampus During Hypoxia. Neuroscience 2018; 392:77-91. [DOI: 10.1016/j.neuroscience.2018.09.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/09/2018] [Accepted: 09/17/2018] [Indexed: 10/28/2022]
|
23
|
Pei Y, Jiao Z, Dong W, Pei L, He X, Wang H, Xu D. Excitotoxicity and compensatory upregulation of GAD67 in fetal rat hippocampus caused by prenatal nicotine exposure are associated with inhibition of the BDNF pathway. Food Chem Toxicol 2018; 123:314-325. [PMID: 30389584 DOI: 10.1016/j.fct.2018.10.062] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/04/2018] [Accepted: 10/29/2018] [Indexed: 10/28/2022]
Abstract
Prenatal nicotine exposure (PNE) can cause hypersensitivity of hypothalamic-pituitary-adrenal (HPA) axis in offspring with intrauterine growth retardation. The purpose of this study was to explore the original mechanism of intrauterine development that mediates hypersensitivity of the HPA axis in offspring due to PNE. Pregnant Wistar rats were injected subcutaneously with 2 mg/kg·d of nicotine on the 9th to the 20th gestational day (GD9-GD20) and the fetuses were extracted at GD20. Compared with the control group, fetal rats by PNE showed increased hippocampal apoptosis, reduced synaptic plasticity and downregulation of the brain-derived neurotrophic factor (BDNF) pathway, whereas glutamic acid decarboxylase 67 (GAD67) expression was upregulated. Rat fetal hippocampal H19-7/IGF1R cell lines were treated with different concentrations of nicotine (1, 10 and 100 μM) for 3 days, the extracellular fluid glutamate (Glu) level increased and similar effects were observed as in vivo. Intervention treatments caused the opposite results. These results indicated that PNE downregulates the BDNF pathway and mediates the hippocampal excitotoxicity; then, the compensatory upregulation of GAD67 causes the imbalance of signal output in the fetal hippocampus. The negative feedback regulation of the paraventricular hypothalamic nucleus by the hippocampus is unbalanced, eventually causing hypersensitivity of the HPA axis of the offspring.
Collapse
Affiliation(s)
- Yun Pei
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Zhexiao Jiao
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Wanting Dong
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Linguo Pei
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Xia He
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Dan Xu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
24
|
Pottoo FH, Tabassum N, Javed MN, Nigar S, Rasheed R, Khan A, Barkat MA, Alam MS, Maqbool A, Ansari MA, Barreto GE, Ashraf GM. The Synergistic Effect of Raloxifene, Fluoxetine, and Bromocriptine Protects Against Pilocarpine-Induced Status Epilepticus and Temporal Lobe Epilepsy. Mol Neurobiol 2018; 56:1233-1247. [PMID: 29881945 DOI: 10.1007/s12035-018-1121-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/11/2018] [Indexed: 10/14/2022]
|
25
|
Shalaby HN, El-Tanbouly DM, Zaki HF. Topiramate mitigates 3-nitropropionic acid-induced striatal neurotoxicity via modulation of AMPA receptors. Food Chem Toxicol 2018; 118:227-234. [PMID: 29753867 DOI: 10.1016/j.fct.2018.05.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 01/10/2023]
Abstract
Prevalence of glutamate receptor subunit 2 (GluR2)-lacking alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors is a hallmark of excitotoxicity-related neurodegenerative diseases. Topiramate (TPM) is a structurally novel anticonvulsant with a well-known modulatory effects on AMPA/kainate subtypes of glutamate receptors. The present study aimed at investigating the neuroprotective potential of TPM on 3-nitropropionic acid (3-NP)-induced striatal neurodegeneration and Huntington's disease-like symptoms. Rats were injected with 3-NP (10 mg/kg/i.p.) for 14 days. TPM (50 mg/kg/p.o.) was given once a day, 1 h before 3-NP. TPM amended 3-NP induced changes in neurobehavioral performance, striatal neurotransmitters levels and histopathological injury. 3-NP control rats showed a significant ablation in the mRNA expression of Ca2+-impermeable Glu2R subunit along with an elevation in its regulatory protein (protein interacting with C kinase-1) PICK1, an effect that was largely reversed by TPM. TPM in addition, enhanced the phosphorylation of the protein kinase B/glycogen synthase kinase-3β/cAMP response element binding protein (Akt/GSK-3β/CREB) cue. Moreover, improvement in oxidative status, suppression of caspase-3 activity and restoration of striatal BDNF were noticed following treatment with TPM. The current study revealed that TPM boosted the neuroprotective (Akt/GSK-3β/CREB) pathway by its negative modulatory effect on AMPA glutamate receptors as well as its direct antioxidant property.
Collapse
Affiliation(s)
- Heba N Shalaby
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalia M El-Tanbouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
26
|
Fouad IA, Sharaf NM, Abdelghany RM, El Sayed NSED. Neuromodulatory Effect of Thymoquinone in Attenuating Glutamate-Mediated Neurotoxicity Targeting the Amyloidogenic and Apoptotic Pathways. Front Neurol 2018; 9:236. [PMID: 29706929 PMCID: PMC5908889 DOI: 10.3389/fneur.2018.00236] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/26/2018] [Indexed: 11/13/2022] Open
Abstract
Overexposure of the glutamatergic N-methyl-d-aspartate (NMDA) receptor to the excitatory neurotransmitter l-glutamic acid leads to neuronal cell death by excitotoxicity as a result of increased intracellular Ca2+, mitochondrial dysfunction, and apoptosis. Moreover, it was previously reported that prolonged activation of the NMDA receptor increased beta-amyloid (Aβ) levels in the brain. Thymoquinone (TQ), the active constituent of Nigella sativa seeds, has been shown to have potent antioxidant and antiapoptotic effects. The aim of the present study was to explore the neuromodulatory effects of different doses of TQ (2.5 and 10 mg/kg) against apoptotic cell death and Aβ formation resulting from glutamate administration in rats using vitamin E as a positive control. Behavioral changes were assessed using Y-maze and Morris water maze tests for evaluating spatial memory and cognitive functions. Caspase-3, Lactate dehydrogenase, Aβ-42, and cytochrome c gene expression were determined. TQ-treated groups showed significant decreases in the levels of all tested biochemical and behavioral parameters compared with the glutamate-treated group. These findings demonstrated that TQ has a promising neuroprotective activity against glutamate-induced neurotoxicity and this effect is mediated through its anti-amyloidogenic, antioxidant, and antiapoptotic activities.
Collapse
Affiliation(s)
- Ibram Amin Fouad
- Department of Pharmacology and Toxicology, German University in Cairo, New Cairo, Egypt
| | - Nadia Mohamed Sharaf
- Department of Pharmacology and Toxicology, German University in Cairo, New Cairo, Egypt
| | | | | |
Collapse
|
27
|
Varma A, Moore MB, Miller CWT, Himelhoch S. Topiramate as Monotherapy or Adjunctive Treatment for Posttraumatic Stress Disorder: A Meta-Analysis. J Trauma Stress 2018; 31:125-133. [PMID: 29388709 DOI: 10.1002/jts.22251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 06/27/2017] [Accepted: 08/10/2017] [Indexed: 11/10/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a chronic and debilitating condition for which clinicians sometimes turn to anticonvulsants as a treatment for symptoms. This study was a systematic review and meta-analysis of randomized controlled trials (RCT) that have assessed the efficacy of topiramate as monotherapy or adjunctive therapy, compared to placebo, for the treatment of PTSD in adults. Prescribers may be reluctant to turn to topiramate, given the commonly reported side effects of impaired cognition, sedation, fatigue, and headache. We searched PubMed, PsycInfo, and Cochrane Central databases for relevant trials. Five studies were identified as RCTs and thus met inclusion criteria; one additional nonpublished study was identified via phone contact with its authors. Of these six studies, one was excluded from the statistical meta-analysis due to its high dropout rate (16 of 40 participants). One of these studies was excluded from a stratified analysis of symptom types because this subscale data were unavailable. For overall symptomatology, topiramate showed a medium, but not significant effect, standardized mean difference (SMD) = 0.55, p = .082. Topiramate showed a small and significant reduction of hyperarousal symptoms, SMD = 0.35, 95% CI [0.029, 0.689], p = .033. Topiramate did not significantly reduce reexperiencing symptoms, SMD = 0.29, 95% CI [-0.019, 0.597], p = .067, or avoidance symptoms, SMD = 0.20, 95% CI [-0.105, 0.509], p = .198. Results did not differ significantly between veteran and nonveteran subjects, or between topiramate as monotherapy and adjunctive therapy. Further studies on topiramate will clarify its role in PTSD treatment.
Collapse
Affiliation(s)
- Archana Varma
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Michael B Moore
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Christopher W T Miller
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Seth Himelhoch
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Rivera-Carvantes MC, Jarero-Basulto JJ, Feria-Velasco AI, Beas-Zárate C, Navarro-Meza M, González-López MB, Gudiño-Cabrera G, García-Rodríguez JC. Changes in the expression level of MAPK pathway components induced by monosodium glutamate-administration produce neuronal death in the hippocampus from neonatal rats. Neuroscience 2017; 365:57-69. [PMID: 28954212 DOI: 10.1016/j.neuroscience.2017.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 09/12/2017] [Accepted: 09/17/2017] [Indexed: 11/18/2022]
Abstract
Excessive Glutamate (Glu) release may trigger excitotoxic cellular death by the activation of intracellular signaling pathways that transduce extracellular signals to the cell nucleus, which determines the onset of a death program. One such signaling pathway is the mitogen-activated protein kinases (MAPK), which is involved in both survival and cell death. Experimental evidences from the use of specific inhibitors supports the participation of some MAPK pathway components in the excitotoxicity mechanism, but the complete process of this activation, which terminates in cell damage and death, is not clearly understood. The present work, we investigated the changes in the expression level of some MAPK-pathway components in hippocampal excitotoxic cell death in the neonatal rats using an experimental model of subcutaneous monosodium glutamate (MSG) administration on postnatal days (PD) 1, 3, 5 and 7. Data were collected at different ages through PD 14. Cell viability was evaluated using fluorescein diacetate mixed with propidium iodide (FDA-PI), and the Nissl-staining technique was used to evaluate histological damage. Transcriptional changes were also investigated in 98 components of the MAPK pathway that are associated with cell damage. These results are an evidence of that repetitive use of MSG, in neonatal rats, induces cell damage-associated transcriptional changes of MAPK components, that might reflect a differential stage of both biochemical and molecular brain maturation. This work also suggests that some of the proteins evaluated such as phosphorylated retinoblastoma (pRb) protein, which was up-regulated, could regulate the response to excitotoxic through modulation of the process of re-entry into the cell cycle in the hippocampus of rats treated with MSG.
Collapse
Affiliation(s)
- Martha Catalina Rivera-Carvantes
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico.
| | - José Jaime Jarero-Basulto
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Alfredo Ignacio Feria-Velasco
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Carlos Beas-Zárate
- Regeneration and Neural Development Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Mónica Navarro-Meza
- Department of Health and Wellness, CUSur, University of Guadalajara, Ciudad Guzman, Jal., Mexico
| | - Mariana Berenice González-López
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Graciela Gudiño-Cabrera
- Regeneration and Neural Development Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | | |
Collapse
|
29
|
Ye X, Yu L, Zuo D, Zhang L, Zu J, Hu J, Tang J, Bao L, Cui C, Zhang R, Jin G, Zan K, Zhang Z, Yang X, Shi H, Zhang Z, Xiao Q, Liu Y, Xiang J, Zhang X, Cui G. Activated mGluR5 protects BV2 cells against OGD/R induced cytotoxicity by modulating BDNF-TrkB pathway. Neurosci Lett 2017. [PMID: 28642149 DOI: 10.1016/j.neulet.2017.06.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Activated Metabotropic glutamate receptors 5(mGluR5) exhibits protective effects against ischemic brain damage, but the underlying mechanisms are not clearly known. Brain-derived neurotrophic factor (BDNF), as a valuable member of neurotrophic factor family, exerts its protection by combining with its high-affinity receptor tyrosine protein kinase B (TrkB). To investigate the role of activated mGluR5 against oxygen-glucose deprivation (OGD)/reoxygenation (R)-mediated cytotoxicity, the cell viability, apoptosis, the release of inflammatory cytokines and accumulation of reactive oxygen species (ROS) were evaluated in BV2 cells (Microglia cell line) with or without OGD/R exposure. Our data show that CHPG (the selective mGluR5 agonist) pretreatment, as an mGluR5 agonist, protected BV2 cells against OGD/R-induced cytotoxicity, apoptosis, the release of inflammatory cytokines, and the accumulation of ROS. However, these effects were significantly reversed by the mGluR5 antagonist MPEP pretreatment. Our data also show that the expressions of BDNF and TrkB were significantly decreased in BV2 cells with OGD/R exposure. CHPG pretreatment significantly enhanced the expressions of BDNF and TrkB in BV2 cells with OGD/R exposure. However, the increased expressions were significantly abrogated by MPEP pretreatment. In addition, inhibition of BDNF/TrKB pathway by K252a also attenuated the protective effects of activated mGluR5 against OGD/R-induced cytotoxicity, apoptosis and the release of inflammatory cytokines. Morever, pretreatment with exogenous BDNF protected BV2 cells against OGD/R induced apoptosis and release of inflammatory cytokines. These data suggested that BDNF/TrKB pathway may be involved in regulating activated mGluR5' protective effects against OGD/R induced cytotoxicity in BV2 cells.
Collapse
Affiliation(s)
- Xinchun Ye
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Lu Yu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Dandan Zuo
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Liang Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Jie Zu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Jinxia Hu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Jiao Tang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Lei Bao
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Chengcheng Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Ruixue Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Guoliang Jin
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Kun Zan
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Zuohui Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Xinxin Yang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Hongjuan Shi
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Zunsheng Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Qihua Xiao
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Yonghai Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Jie Xiang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Xueling Zhang
- Department of Neurology, Suqian People's Hospital, Suqian, Jiangsu Province, 223800, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China.
| |
Collapse
|
30
|
Motaghinejad M, Motevalian M, Babalouei F, Abdollahi M, Heidari M, Madjd Z. Possible involvement of CREB/BDNF signaling pathway in neuroprotective effects of topiramate against methylphenidate induced apoptosis, oxidative stress and inflammation in isolated hippocampus of rats: Molecular, biochemical and histological evidences. Brain Res Bull 2017; 132:82-98. [PMID: 28552672 DOI: 10.1016/j.brainresbull.2017.05.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 05/13/2017] [Accepted: 05/19/2017] [Indexed: 12/29/2022]
Abstract
Chronic abuse of methylphenidate (MPH) can cause serious neurotoxicity. The neuroprotective effects of topiramate (TPM) were approved, but its putative mechanism remains unclear. In current study the role of CREB/BDNF signaling pathway in TPM protection against methylphenidate-induced neurotoxicity in rat hippocampus was evaluated. 60 adult male rats were divided randomly into six groups. Groups received MPH (10mg/kg) only and concurrently with TPM (50mg/kg and 100mg/kg) and TPM (50 and 100mg/kg) only for 14 days. Open field test (OFT) was used to investigate motor activity. Some biomarkers of apoptotic, anti-apoptotic, oxidative, antioxidant and inflammatory factors were also measured in hippocampus. Expression of total (inactive) and phosphorylated (active) CREB and BDNF were also measured in gene and protein levels in dentate gyrus (DG) and CA1 areas of hippocampus. MPH caused significant decreases in motor activity in OFT while TPM (50 and 100mg/kg) inhibited MPH-induced decreases in motor activity. On the other hand, MPH caused remarkable increases in Bax protein level, lipid peroxidation, catalase activity, IL-1β and TNF-α levels in hippocampal tissue. MPH also caused significant decreases of superoxide dismutase, activity and also decreased CREB, in both forms, BDNF and Bcl-2 protein levels. TPM, by the mentioned doses, attenuated these effects and increased superoxide dismutase, glutathione peroxidase and glutathione reductase activities and also increased CREB, in both forms, BDNF and Bcl-2 protein levels and inhibited MPH induced increase in Bax protein level, lipid peroxidation, catalase activity, IL-1β and TNF-α levels. TPM also inhibited MPH induced decreases in cell number and changes in cell shapes in DG and CA1 areas. TPM can probably act as a neuroprotective agent against MPH induced neurotoxicity and this might have been mediated by CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Manijeh Motevalian
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Babalouei
- Deparemten of Chemistry, Faculty of Science, Islamic Azad University, Share-Qods Brach, Tehran, Iran
| | - Mohammad Abdollahi
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansour Heidari
- Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center and Department of Pathology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Can Co-Activation of Nrf2 and Neurotrophic Signaling Pathway Slow Alzheimer's Disease? Int J Mol Sci 2017; 18:ijms18061168. [PMID: 28561773 PMCID: PMC5485992 DOI: 10.3390/ijms18061168] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/22/2017] [Accepted: 05/27/2017] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifaceted disease that is hard to treat by single-modal treatment. AD starts with amyloid peptides, mitochondrial dysfunction, and oxidative stress and later is accompanied with chronic endoplasmic reticulum (ER) stress and autophagy dysfunction, resulting in more complicated pathogenesis. Currently, few treatments can modify the complicated pathogenic progress of AD. Compared to the treatment with exogenous antioxidants, the activation of global antioxidant defense system via Nrf2 looks more promising in attenuating oxidative stress in AD brains. Accompanying the activation of the Nrf2-mediated antioxidant defense system that reduce the AD-causative factor, oxidative stress, it is also necessary to activate the neurotrophic signaling pathway that replaces damaged organelles and molecules with new ones. Thus, the dual actions to activate both the Nrf2 antioxidant system and neurotrophic signaling pathway are expected to provide a better strategy to modify AD pathogenesis. Here, we review the current understanding of AD pathogenesis and neuronal defense systems and discuss a possible way to co-activate the Nrf2 antioxidant system and neurotrophic signaling pathway with the hope of helping to find a better strategy to slow AD.
Collapse
|
32
|
Motaghinejad M, Motevalian M, Abdollahi M, Heidari M, Madjd Z. Topiramate Confers Neuroprotection Against Methylphenidate-Induced Neurodegeneration in Dentate Gyrus and CA1 Regions of Hippocampus via CREB/BDNF Pathway in Rats. Neurotox Res 2017; 31:373-399. [PMID: 28078543 DOI: 10.1007/s12640-016-9695-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/21/2016] [Accepted: 12/26/2016] [Indexed: 12/18/2022]
Abstract
Methylphenidate (MPH) abuse can cause serious neurological damages. The neuroprotective effects of topiramate (TPM) have been reported already, but its mechanism of action still remains unclear. The current study evaluates in vivo role of CREB/BDNF in TPM protection of the rat hippocampal cells from methylphenidate-induced apoptosis, oxidative stress, and inflammation. A total of 60 adult male rats were divided into six groups. Groups 1 and 2 received normal saline (0.7 ml/rat) and MPH (10 mg/kg) respectively for 14 days. Groups 3 and 4 were concurrently treated with MPH (10 mg/kg) and TPM 50 and 100 mg/kg respectively for 14 days. Groups 5 and 6 were treated with 50 and 100 mg/kg TPM only respectively. After drug administration, open field test (OFT) was used to investigate motor activity. The hippocampus was then isolated and the apoptotic, antiapoptotic, oxidative, antioxidant, and inflammatory factors were measured. Expression of the total and phosphorylated CREB and BDNF in gene and protein levels, and gene expression of Ak1, CaMK4, MAPK3, PKA, and c-Fos levels were also measured. MPH significantly decreased motor activity in OFT. TPM (50 and 100 mg/kg) decreased MPH-induced motor activity disturbance. Additionally, MPH significantly increased Bax protein level, CaMK4 gene expression, lipid peroxidation, catalase activity, mitochondrial GSH, IL-1β, and TNF-α levels in isolated hippocampal cells. Also CREB, in total and phosphorylated forms, BDNF and Bcl-2 protein levels, Ak1, MAPK3, PKA and c-Fos gene expression, superoxide dismutase, glutathione peroxidase, and glutathione reductase activities decreased significantly by MPH. TPM (50 and 100 mg/kg), both in the presence and absence of MPH, attenuated the effects of MPH. Immunohistochemistry data showed that TPM increased localization of the total and phosphorylated forms of CREB in dentate gyrus (DG) and CA1 areas of the hippocampus. It seems that TPM can be used as a neuroprotective agent against apoptosis, oxidative stress, and neuroinflammation induced by frequent use of MPH. This might be probably mediated by the CREB/BDNF and their upstream signaling pathways.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat high way, Beside Milad Tower, Tehran, 14496-14525, Iran
| | - Manijeh Motevalian
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat high way, Beside Milad Tower, Tehran, 14496-14525, Iran.
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansour Heidari
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center and Department of pathology, Faculty of medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Jia Y, Dong Z, Yan T, Wu B, Liao Z, Bi K, Gong P, Suna B. Antidepressant-like activity of red wine phenolic extracts in repeated corticosterone-induced depression mice via BDNF/TrkB/CREB signaling pathway. BIO WEB OF CONFERENCES 2016. [DOI: 10.1051/bioconf/20160704009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
34
|
Neuroprotective effects of various doses of topiramate against methylphenidate-induced oxidative stress and inflammation in isolated rat amygdala: the possible role of CREB/BDNF signaling pathway. J Neural Transm (Vienna) 2016; 123:1463-1477. [PMID: 27665547 DOI: 10.1007/s00702-016-1619-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 09/05/2016] [Indexed: 12/26/2022]
Abstract
Methylphenidate (MPH) abuse damages brain cells. The neuroprotective effects of topiramate (TPM) have been reported previously, but its exact mechanism of action still remains unclear. This study investigated the in vivo role of various doses of TPM in the protection of rat amygdala cells against methylphenidate-induced oxidative stress and inflammation. Seventy adult male rats were divided into seven groups. Groups 1 and 2 received normal saline (0.7 ml/rat) and MPH (10 mg/kg), respectively, for 21 days. Groups 3, 4, 5, 6, and 7 were concurrently treated with MPH (10 mg/kg) and TPM (10, 30, 50, 70, and 100 mg/kg), respectively, for 21 days. elevated plus maze (EPM) was used to assess motor activity disturbances. In addition, oxidative, antioxidantand inflammatory factors and CREB, Ak1, CAMK4, MAPK3, PKA, BDNF, and c FOS gene levels were measured by RT-PCR, and also, CREB and BDNF protein levels were measured by WB in isolated amygdalae. MPH significantly disturbed motor activity and TPM (70 and 100 mg/kg) neutralized its effects. MPH significantly increased lipid peroxidation, mitochondrial GSSG levels and IL-1β and TNF-α level and CAMK4 gene expression in isolated amygdala cells. In contrast, superoxide dismutase, glutathione peroxidase, and glutathione reductase activities and CREB, BDNF Ak1, MAPK3, PKA, BDNF, and c FOS expression significantly decreased. The various doses of TPM attenuated these effects of MPH. It seems that TPM can be used as a neuroprotective agent and is a good candidate against MPH-induced neurodegeneration.
Collapse
|
35
|
Ji LL, Peng JB, Fu CH, Cao D, Li D, Tong L, Wang ZY. Activation of Sigma-1 receptor ameliorates anxiety-like behavior and cognitive impairments in a rat model of post-traumatic stress disorder. Behav Brain Res 2016; 311:408-415. [PMID: 27275520 DOI: 10.1016/j.bbr.2016.05.056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/21/2016] [Accepted: 05/26/2016] [Indexed: 11/25/2022]
Abstract
Among learning and memory processes, fear memories are crucial in some psychiatric disorders like post-traumatic stress disorder (PTSD). Accumulating evidence shows that the sigma-1 receptor (Sig-1R) has comprehensive involvement in cognitive impairment and neuroprotective effects. It has also been reported that BDNF appears to enhance extinction of fear in anxiety disorders via the MAPK signaling cascade. However, it remains unclear whether BDNF-TrkB-MAPK pathway may be mechanistically involved in the therapeutic effect of sigma-1 receptor in the development of PTSD. To address this question, rats were subjected to a classical single-prolonged stress procedure (SPS) and kept undisturbed for 7 days. After that, rats were re-stressed by re-exposure to the forced swim component of SPS (RSPS). Behavior tests were subsequently performed to assess anxiety and cognitive impairments. Furthermore, we analyzed the expression of BDNF and the phosphorylation of TrkB and three MAPK pathways, namely, the ERK, JNK and p38. We found that the levels of BDNF and p-TrkB were increased following the RSPS procedure, which were reversed by the administration of PRE-084. Meanwhile, among the three MAPK signaling pathways, only the p-ERK expression was increased following the RSPS procedure. Collectively, our results indicate that BDNF-TrkB-ERK signaling pathway may be involved in the activation of sigma-1 receptor to yield therapeutic benefits for PTSD.
Collapse
Affiliation(s)
- Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Jun-Bo Peng
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Dong Cao
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Dan Li
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China.
| | - Zhen-Yu Wang
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
36
|
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 DOI: 10.1007/s10571-015-0276-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [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
|
37
|
Protective effect of liquiritigenin on depressive-like behavior in mice after lipopolysaccharide administration. Psychiatry Res 2016; 240:131-136. [PMID: 27107388 DOI: 10.1016/j.psychres.2016.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/27/2016] [Accepted: 04/01/2016] [Indexed: 12/16/2022]
Abstract
Liquiritigenin (Liq), the main active ingredient of traditional Chinese medicine licorice, possesses anti-inflammatory and neuroprotective properties. The current investigation was designed to explore whether liquiritigenin could relieve lipopolysaccharide (LPS)-induced depression-like behavior in mice and the underlying mechanism. Liquiritigenin (7.5mg/kg, 15mg/kg) and fluoxetine (20mg/kg) were pretreated intragastrically once daily for 7 consecutive days. LPS (0.5mg/kg) was injected subcutaneously to establish the depression model 30min after pretreatment on day 7. Interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels in serum and hippocampus were detected by enzyme-linked immunosorbent assay (ELISA). Behavioral assessment was conduct 24h post LPS injection. The expressions of p65NF-κB, IκBα, brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase B (TrkB) in hippocampus were determined by western blot. The obtained results showed that liquiritigenin effectively reduced the levels of pro-inflammatory cytokines and the expressions of p-p65NF-κB and p-IκBα. Furthermore, liquiritigenin preconditioning could down-regulate the immobility time in tail suspension test (TST), forced swimming test (FST) and up-regulate BDNF and TrkB contents in hippocampus. Thus, it is assumed that the antidepressant activity of liquiritigenin might be attributed to its anti-inflammatory property and BDNF/TrkB signaling pathway.
Collapse
|
38
|
Erythropoietin Pathway: A Potential Target for the Treatment of Depression. Int J Mol Sci 2016; 17:ijms17050677. [PMID: 27164096 PMCID: PMC4881503 DOI: 10.3390/ijms17050677] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 04/05/2016] [Accepted: 04/27/2016] [Indexed: 12/21/2022] Open
Abstract
During the past decade, accumulating evidence from both clinical and experimental studies has indicated that erythropoietin may have antidepressant effects. In addition to the kidney and liver, many organs have been identified as secretory tissues for erythropoietin, including the brain. Its receptor is expressed in cerebral and spinal cord neurons, the hypothalamus, hippocampus, neocortex, dorsal root ganglia, nerve axons, and Schwann cells. These findings may highlight new functions for erythropoietin, which was originally considered to play a crucial role in the progress of erythroid differentiation. Erythropoietin and its receptor signaling through JAK2 activate multiple downstream signaling pathways including STAT5, PI3K/Akt, NF-κB, and MAPK. These factors may play an important role in inflammation and neuroprogression in the nervous system. This is particularly true for the hippocampus, which is possibly related to learning, memory, neurocognitive deficits and mood alterations. Thus, the influence of erythropoietin on the downstream pathways known to be involved in the treatment of depression makes the erythropoietin-related pathway an attractive target for the development of new therapeutic approaches. Focusing on erythropoietin may help us understand the pathogenic mechanisms of depression and the molecular basis of its treatment.
Collapse
|
39
|
Osborne DM, Fitzgerald DP, O'Leary KE, Anderson BM, Lee CC, Tessier PM, McNay EC. Intrahippocampal administration of a domain antibody that binds aggregated amyloid-β reverses cognitive deficits produced by diet-induced obesity. Biochim Biophys Acta Gen Subj 2016; 1860:1291-8. [PMID: 26970498 DOI: 10.1016/j.bbagen.2016.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/20/2016] [Accepted: 03/06/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND The prevalence of high fat diets (HFD), diet-induced obesity (DIO) and Type 2 diabetes continues to increase, associated with cognitive impairment in both humans and rodent models. Mechanisms transducing these impairments remain largely unknown: one possibility is that a common mechanism may be involved in the cognitive impairment seen in obese and/or diabetic states and in dementia, specifically Alzheimer's disease (AD). DIO is well established as a risk factor for development of AD. Oligomeric amyloid-β (Aβ) is neurotoxic, and we showed that intrahippocampal oligomeric Aβ produces cognitive and metabolic dysfunction similar to that seen in DIO or diabetes. Moreover, animal models of DIO show elevated brain Aβ, a hallmark of AD, suggesting that this may be one source of cognitive impairment in both conditions. METHODS Intrahippocampal administration of a novel anti-Aβ domain antibody for aggregated Aβ, or a control domain antibody, to control or HFD-induced DIO rats. Spatial learning measured in a conditioned contextual fear (CCF) task after domain antibody treatment; postmortem, hippocampal NMDAR and AMPAR were measured. RESULTS DIO caused impairment in CCF, and this impairment was eliminated by intrahippocampal administration of the active domain antibody. Measurement of hippocampal proteins suggests that DIO causes dysregulation of hippocampal AMPA receptors, which is also reversed by acute domain antibody administration. CONCLUSIONS Our findings support the concept that oligomeric Aβ within the hippocampus of DIO animals may not only be a risk factor for development of AD but may also cause cognitive impairment before the development of dementia. GENERAL SIGNIFICANCE AND INTEREST Our work integrates the engineering of domain antibodies with conformational- and sequence-specificity for oligomeric amyloid beta with a clinically relevant model of diet-induced obesity in order to demonstrate not only the pervasive effects of obesity on several aspects of brain biochemistry and behavior, but also the bioengineering of a successful treatment against the long-term detrimental effects of a pre-diabetic state on the brain. We show for the first time that cognitive impairment linked to obesity and/or insulin resistance may be due to early accumulation of oligomeric beta-amyloid in the brain, and hence may represent a pre-Alzheimer's state.
Collapse
Affiliation(s)
- Danielle M Osborne
- Behavioral Neuroscience, University at Albany, Albany, NY, United States; Center for Neuroscience Research, University at Albany, Albany, NY, United States
| | - Dennis P Fitzgerald
- Hofstra North Shore-Long Island School of Medicine, Hofstra University, Hempstead, NY, United States
| | - Kelsey E O'Leary
- University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Brian M Anderson
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY, United States
| | - Christine C Lee
- Center for Biotechnology and Interdisciplinary Studies, Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Peter M Tessier
- Center for Biotechnology and Interdisciplinary Studies, Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Ewan C McNay
- Behavioral Neuroscience, University at Albany, Albany, NY, United States; Center for Neuroscience Research, University at Albany, Albany, NY, United States; Biological Sciences, University at Albany, Albany, NY, United States.
| |
Collapse
|
40
|
Yuan L, Liu J, Dong R, Zhu J, Tao C, Zheng R, Zhu S. 14,15-epoxyeicosatrienoic acid promotes production of brain derived neurotrophic factor from astrocytes and exerts neuroprotective effects during ischaemic injury. Neuropathol Appl Neurobiol 2016; 42:607-620. [PMID: 26526810 DOI: 10.1111/nan.12291] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 10/09/2015] [Accepted: 10/27/2015] [Indexed: 02/06/2023]
Abstract
AIMS 14,15-Epoxyeicosatrienoic acid (14,15-EET) is abundantly expressed in brain and exerts protective effects against ischaemia. 14,15-EET is hydrolysed by soluble epoxide hydrolase (sEH). sEH-/- mice show a higher level of 14,15-EET in the brain. Astrocytes play a pivotal role in neuronal survival under ischaemic conditions. However, it is unclear whether the neuroprotective effect of 14,15-EET is associated with astrocytes. METHODS A mouse model of focal cerebral ischaemia was induced by middle cerebral artery occlusion. Oxygen-glucose deprivation/reoxygenation (OGD/R) was performed on cultured murine astrocytes, neurons and a human cell line. Cell viabilities were measured by 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide (MTT) assay. The mRNA expressions were quantified by real-time PCR. Brain derived neurotrophic factor (BDNF) concentration was measured by ELISA. Protein expressions were quantified by Western blotting. BDNF and peroxisome proliferators-activated receptor gamma (PPAR-γ) expressions were analysed by confocal microscopy. RESULTS Decreased infarct volumes, elevated BDNF expression and increased numbers of BDNF/GFAP Glial Fibrillary Acidic Protein double-positive cells were observed in the ischaemic penumbra of sEH-/- mice. The decreased infarct volumes of sEH-/- mice were diminished by intracerebroventricular injection of a blocker of BDNF receptor. 14,15-EET increases BDNF expression and cell viability of murine astrocytes and U251 cells by BDNF-TrkB Tyrosine receptor kinase-B-extracellular signal-regulated kinase 1/2 signalling during OGD/R. 14,15-EET protects neurons from OGD/R by stimulating the production of astrocyte-derived BDNF. 14,15-EET stimulates the production of astrocyte-derived BDNF through PPAR-γ/p-cAMP-response element binding protein signal pathways. CONCLUSIONS Our study demonstrates the importance of 14,15-EET-mediated production of astrocyte-derived BDNF for enhancing viability of astrocytes and protecting neurons from the ischaemic injury and provides insights into the mechanism by which 14,15-EET is involved in neuroprotection.
Collapse
Affiliation(s)
- L Yuan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - J Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - R Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - J Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - C Tao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - R Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - S Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
41
|
Gao S, Li W, Zou W, Zhang P, Tian Y, Xiao F, Gu H, Tang X. H2S protects PC12 cells against toxicity of corticosterone by modulation of BDNF-TrkB pathway. Acta Biochim Biophys Sin (Shanghai) 2015; 47:915-24. [PMID: 26423115 DOI: 10.1093/abbs/gmv098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/24/2015] [Indexed: 12/31/2022] Open
Abstract
Corticosterone, one of the glucocorticoids, is toxic to neurons and plays an important role in depressive-like behavior and depression. We previously showed that hydrogen sulfide (H2S), a novel physiological mediator, plays an inhibitory role in depression. However, the mechanism underlying H2S-triggered antidepressant-like role is not clearly known. Brain-derived neurotrophic factor (BDNF), a neurotrophic factor, plays a neuroprotective role that is mediated by its high-affinity tropomysin-related kinase B (TrkB) receptor. In this study, to investigate the underlying mechanism of H2S-induced antidepressant-like role, we explored whether H2S could protect neurons against corticosterone-mediated cyctotoxicity and whether this protective role of H2S was involved in the regulation of BDNF-TrkB pathway. Our data demonstrated that sodium hydrosulfide (NaHS), the donor of H2S, could prevent corticosterone-induced cytotoxicity, apoptosis, accumulation of intracellular reactive oxygen species (ROS) and loss of mitochondrial membrane potential (MMP) in PC12 cells. NaHS not only induced the up-regulation of BDNF but also prevented the down-regulation of BDNF by corticosterone. It was also found that blocking BDNF-TrkB pathway by K252a, an inhibitor of TrkB, abolished the protection of H2S against corticosterone-induced cytotoxicity, apoptosis, accumulation of ROS, and loss of MMP. These results suggest that H2S protects against the neurotoxicity of corticosterone by modulation of the BDNF-TrkB pathway.
Collapse
Affiliation(s)
- Shenglan Gao
- Institute of Neuroscience, Medical College, University of South China, Hengyang 42100, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| | - Wenting Li
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China
| | - Wei Zou
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China
| | - Ping Zhang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China
| | - Ying Tian
- Department of Biochemistry, Medical College, University of South China, Hengyang 421001, China
| | - Fan Xiao
- Institute of Neuroscience, Medical College, University of South China, Hengyang 42100, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| | - Hongfeng Gu
- Institute of Neuroscience, Medical College, University of South China, Hengyang 42100, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China
| | - Xiaoqing Tang
- Institute of Neuroscience, Medical College, University of South China, Hengyang 42100, China Key Laboratory for Cognitive Disorders and Neurodegenerative Diseases, University of South China, Hengyang 421001, China Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang 421001, China
| |
Collapse
|