1
|
Moosavi M, Soukhaklari R, Bagheri-Mohammadi S, Firouzan B, Javadpour P, Ghasemi R. Nanocurcumin prevents memory impairment, hippocampal apoptosis, Akt and CaMKII-α signaling disruption in the central STZ model of Alzheimer's disease in rat. Behav Brain Res 2024; 471:115129. [PMID: 38942084 DOI: 10.1016/j.bbr.2024.115129] [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: 05/06/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
The central route of streptozotocin (STZ) administration has been introduced as a rat model of sporadic Alzheimer's disease (AD). Curcumin was suggested to possess possible neuroprotective effects, which may be profitable in AD. However, the low bioavailability of curcumin hinders its beneficial effects in clinical studies. Earlier studies suggested that a bovine serum albumin-based nanocurcumin, produces superior neuroprotective effects compared to natural curcumin. In the present study, the protective effect of nanocurcumin in rat model of central STZ induced memory impairment was assessed. In addition, due to the importance of the hippocampus in memory, the amounts of hippocampal active caspase-3, Akt, and CaMKII-α were evaluated. Adult male Wistar rats weighing 250-300 g were used. STZ (icv) was injected during days 1 and 3 (3 mg/kg in divided), and nanocurcumin or curcumin 50 mg/kg/oral gavage was administered daily during days 4-14. Morris water maze training was performed on days 15-17, and the retention memory test was achieved on the 18th day. Following memory assessment, the rats were sacrificed and the hippocampi were used to assess caspase-3 cleavage, Akt, and CaMKII-α signaling. The findings revealed that nanocurcumin ingestion (but not natural curcumin) in the dose of 50 mg/kg was capable to prevent the impairment of water maze learning and memory induced by central STZ. Molecular assessments indicated that STZ treatment increased the caspase-3 cleavage in the hippocampus while deactivating Akt and CaMKII-α. Nanocurcumin reduced caspase-3 cleavage to a non-significant level compared to control group and restored Akt and CaMKII-α within the hippocampus while natural curcumin exerted no significant effect. These findings might suggest that nanocurcumin can restore memory deficit, hippocampal apoptosis as well as Akt and CaMKII-α signaling disruption associated with brain insulin resistance.
Collapse
Affiliation(s)
- Maryam Moosavi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Roksana Soukhaklari
- Shiraz Neuroscience Research Centre, Shiraz University of Medical sciences, Shiraz, Iran; Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Saeid Bagheri-Mohammadi
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Bita Firouzan
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Javadpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Izadi S, Abdolrezaei M, Soukhaklari R, Moosavi M. Memory impairment induced by aluminum nanoparticles is associated with hippocampal IL-1 and IBA-1 upregulation in mice. Neurol Res 2024; 46:284-290. [PMID: 38145565 DOI: 10.1080/01616412.2023.2298137] [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: 06/21/2023] [Accepted: 12/18/2023] [Indexed: 12/27/2023]
Abstract
OBJECTIVES Increasing evidence indicates a link between aluminum (Al) intake and Alzheimer's disease (AD). The main entry of Al into the human body is through oral route, and in the digestive tract, under the influence of the pH change, Al can be transformed into Al nanoparticles (Al-NP). However, studies related to the effect of Al-NP on the brain are limited and need further investigation. Neuro-inflammation is considered as one of the principal features of AD. Microglial activation and expression of the inflammatory cytokine IL-1β (interleukin-1β) in the brain have been used as hallmarks of brain inflammation. Therefore, in the present study, the hippocampal levels of ionized calcium-binding adaptor molecule 1 (IBA-1), as the marker of microglia activation, and IL-1β were assessed. METHODS Adult male NMRI mice were treated with Al-NP (5 or 10 mg/kg) for 5 days. A novel object recognition (NOR) test was used to assess memory. Following cognitive assessments, the hippocampal tissues were isolated to analyze the levels of IL-1β and IBA-1 as well as beta actin proteins using western blot technique. RESULTS Al-NP in both doses of 5 and 10 mg/kg impaired NOR memory in mice. In addition, Al-NP increased IL-1β and IBA-1 in the hippocampus. DISCUSSION These findings indicate that the memory impairing effect of Al-NP coincides with hippocampal inflammation. According to the proposed relationship between AD and Al toxicity, this study can increase the knowledge about the toxic effects of Al-NP and highlight the need to limit the use of this nanoparticle.
Collapse
Affiliation(s)
- Sadegh Izadi
- Clinical Neurology Research Center and Department of Neurology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Abdolrezaei
- Clinical Neurology Research Center and Department of Neurology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roksana Soukhaklari
- Shiraz Neuroscience Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Maryam Moosavi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Ostovan VR, Baberi N, Farokhi MR, Moezi L, Pirsalami F, Soukhaklari R, Moosavi M. Cholinergic deficit induced memory retrieval impairment and hippocampal CaMKII-alpha deregulation is counteracted by sub-chronic agmatine treatment in mice. Neurol Res 2023; 45:1091-1099. [PMID: 37733020 DOI: 10.1080/01616412.2023.2257417] [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: 10/25/2022] [Accepted: 07/02/2023] [Indexed: 09/22/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disease characterized by brain cholinergic dysfunction. Evidence suggests the impairment of memory retrieval phase in AD. It has been shown that CaMKII-α expressing neurons are selectively reduced in the hippocampus in AD brains. The present study aimed to investigate the effect of scopolamine on the memory retrieval phase and the hippocampal CaMKII-α signaling. In addition, the effect of sub-chronic administration of agmatine against scopolamine induced memory and possible hippocampal CaMKII-α deregulation was investigated in mice. Adult male NMRI mice were administered with agmatine at the doses of 5, 10, 20, 30 and 40 mg/kg/i.p. or saline for 11 days. Acquisition and retrieval tests of passive avoidance task were performed on days 10 and 11, respectively (30 Min following agmatine treatment). Scopolamine (1 mg/kg/i.p.) was administered once, 30 Min before retrieval test. Upon completion of the behavioral tasks, the hippocampi were isolated for western blot analysis to detect the phosphorylated and total levels of CaMKII-α and beta actin proteins. The results showed that scopolamine induced memory retrieval deficit and decreased the phosphorylated level of hippocampal CaMKII-α. Sub-chronic agmatine treatment at the dose of 40 mg/kg prevented scopolamine induced memory retrieval deficit and restored the level of hippocampal phosphorylated CaMKII-α. This study suggests that hippocampal CaMKII-α might play a role in scopolamine induced amnesia and sub-chronic agmatine prevents the impairing effect of scopolamine on the retrieval phase of memory and the phosphorylation of hippocampal CaMKII-α protein.
Collapse
Affiliation(s)
- Vahid Reza Ostovan
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nahid Baberi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Reza Farokhi
- Shiraz Neuroscience Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Moezi
- Department of Pharmacology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatema Pirsalami
- Department of Pharmacology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Moosavi
- Nanomedicine and Nanobiology Research Center, Mohammad Rasoolullah (PBUH) Research Tower, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Jiang J, Zhang L, Zou J, Liu J, Yang J, Jiang Q, Duan P, Jiang B. Phosphorylated S6K1 and 4E-BP1 play different roles in constitutively active Rheb-mediated retinal ganglion cell survival and axon regeneration after optic nerve injury. Neural Regen Res 2023; 18:2526-2534. [PMID: 37282486 PMCID: PMC10360084 DOI: 10.4103/1673-5374.371372] [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: 06/08/2023] Open
Abstract
Ras homolog enriched in brain (Rheb) is a small GTPase that activates mammalian target of rapamycin complex 1 (mTORC1). Previous studies have shown that constitutively active Rheb can enhance the regeneration of sensory axons after spinal cord injury by activating downstream effectors of mTOR. S6K1 and 4E-BP1 are important downstream effectors of mTORC1. In this study, we investigated the role of Rheb/mTOR and its downstream effectors S6K1 and 4E-BP1 in the protection of retinal ganglion cells. We transfected an optic nerve crush mouse model with adeno-associated viral 2-mediated constitutively active Rheb and observed the effects on retinal ganglion cell survival and axon regeneration. We found that overexpression of constitutively active Rheb promoted survival of retinal ganglion cells in the acute (14 days) and chronic (21 and 42 days) stages of injury. We also found that either co-expression of the dominant-negative S6K1 mutant or the constitutively active 4E-BP1 mutant together with constitutively active Rheb markedly inhibited axon regeneration of retinal ganglion cells. This suggests that mTORC1-mediated S6K1 activation and 4E-BP1 inhibition were necessary components for constitutively active Rheb-induced axon regeneration. However, only S6K1 activation, but not 4E-BP1 knockdown, induced axon regeneration when applied alone. Furthermore, S6K1 activation promoted the survival of retinal ganglion cells at 14 days post-injury, whereas 4E-BP1 knockdown unexpectedly slightly decreased the survival of retinal ganglion cells at 14 days post-injury. Overexpression of constitutively active 4E-BP1 increased the survival of retinal ganglion cells at 14 days post-injury. Likewise, co-expressing constitutively active Rheb and constitutively active 4E-BP1 markedly increased the survival of retinal ganglion cells compared with overexpression of constitutively active Rheb alone at 14 days post-injury. These findings indicate that functional 4E-BP1 and S6K1 are neuroprotective and that 4E-BP1 may exert protective effects through a pathway at least partially independent of Rheb/mTOR. Together, our results show that constitutively active Rheb promotes the survival of retinal ganglion cells and axon regeneration through modulating S6K1 and 4E-BP1 activity. Phosphorylated S6K1 and 4E-BP1 promote axon regeneration but play an antagonistic role in the survival of retinal ganglion cells.
Collapse
Affiliation(s)
- Jikuan Jiang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Lusi Zhang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Jingling Zou
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Jingyuan Liu
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Jia Yang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Qian Jiang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Peiyun Duan
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| | - Bing Jiang
- Department of Ophthalmology, Second Xiangya Hospital, Central South University; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan Province, China
| |
Collapse
|
5
|
Izadi S, Rezaei A, Esmaili Z, Soukhaklari R, Moosavi M. Agmatine prevents the memory impairment and the dysfunction of hippocampal GSK-3β and ERK signaling induced by aluminum nanoparticle in mice. Behav Pharmacol 2023:00008877-990000000-00046. [PMID: 37401404 DOI: 10.1097/fbp.0000000000000735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
The growing usage of aluminum nanoparticles (Al-NP) and their exposure may influence body function. Considering the proposed relationship between Al and the pathogenesis of Alzheimer's disease and the concern about the effect of this nanoparticle on brain health and cognitive function, the use of neuroprotective agents might be helpful. According to the reported neuroprotective effects of agmatine, in the present study, the possible protective effect of agmatine was assessed in mice model of Al-NP-induced memory impairment. In addition, due to the roles of hippocampal Glycogen synthase kinase-3 beta (GSK-3β) and ERK signaling in memory and its disorders, these pathways were also investigated. Al-NP (10 mg/kg/p.o.) with/without agmatine (5 or 10 mg/kg/i.p.) was administered to adult male NMRI mice for 5 days. Novel object recognition (NOR) test session was used to assess cognitive function. Following the behavioral assessments, the hippocampi were used to determine the phosphorylated and total levels of GSK-3β and ERK as well as GAPDH using western blot analysis. The results showed that Al-NP impaired NOR memory in mice while agmatine 10 mg/kg prevented the memory deficit induced by Al-NP. Furthermore, Al-NP activated GSK-3β as well as ERK signals within the hippocampus while agmatine prevented the effects of Al-NP on GSK-3β and ERK signals within the hippocampus. Besides supporting the neuroprotective effects of agmatine, these findings suggest the possibility of the connection of hippocampal GSK-3β and ERK signaling in the neuroprotective effect of this polyamine against Al-NP.
Collapse
Affiliation(s)
- Sadegh Izadi
- Clinical Neurology Research Center and Department of Neurology, Shiraz University of Medical Sciences
| | - Amin Rezaei
- Clinical Neurology Research Center and Department of Neurology, Shiraz University of Medical Sciences
| | - Zahra Esmaili
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences
| | - Roksana Soukhaklari
- Shiraz Neuroscience Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Maryam Moosavi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences
| |
Collapse
|
6
|
Mao J, Liu P, Han W, Mo R, Guo S, Sun J. The Influence of GFRαl Inhibition on Proliferation and Apoptosis of Spermatogenic Cells. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722060081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
7
|
Farokhi MR, Taherifard E, SoukhakLari R, Moezi L, Pirsalami F, Savardashtaki A, Moosavi M. The memory modulatory effect of agmatine in passive avoidance task coincides with alterations of hippocampal CaMKII-α and ERK signaling in mice. Eur J Pharmacol 2022; 923:174928. [PMID: 35398030 DOI: 10.1016/j.ejphar.2022.174928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 11/03/2022]
Abstract
Agmatine is a polyamine suggested to act as a supposed neurotransmitter in the brain. Evidence has indicated that acute agmatine administration might modulate memory. The present study aimed to investigate the effect of repeated agmatine treatment on passive avoidance memory, hippocampal calcium-calmodulin-dependent protein kinase II-alpha (CaMKII-α), and Extracellular Signal-Regulated Kinase (ERK) signaling pathways in naive mice. Adult male NMRI mice were treated with agmatine (10, 20, 30, 40, and 80 mg/kg/ip) or saline for 11 days. Acquisition and retention tests of passive avoidance memory were performed on days 10 and 11, respectively. Following the memory retention test, the hippocampi were assessed for the levels of CaMKII-α and ERK using the western blotting technique. The results revealed the dose-dependent effect of agmatine on the passive avoidance memory. Accordingly, the memory was impaired in lower doses, but was improved in higher ones. Agmatine in none of the doses affected the nociception of the mice in tail-flick test. Furthermore, agmatine increased the phosphorylation of CaMKII-α and ERK in the hippocampus at memory enhancing doses, while ERK phosphorylation decreased following the impairing doses of agmatine. Thus, the dose-dependent effect of agmatine on memory might be related to its modulatory effect on CaMKII-α and ERK signal transduction, eventually regulating the memory process.
Collapse
Affiliation(s)
- Majid Reza Farokhi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Erfan Taherifard
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Students Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roksana SoukhakLari
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Students Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Moezi
- Department of Pharmacology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran; Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatema Pirsalami
- Department of Pharmacology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Moosavi
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Mehrbeheshti N, Esmaili Z, Ahmadi M, Moosavi M. A dose response effect of oral aluminum nanoparticle on novel object recognition memory, hippocampal caspase-3 and MAPKs signaling in mice. Behav Brain Res 2022; 417:113615. [PMID: 34606775 DOI: 10.1016/j.bbr.2021.113615] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/30/2022]
Abstract
The increasing use of aluminum nanoparticles (nano-Al) leads to increased human exposure and might affect human health. Considering the suggested connection between aluminum exposure and Alzheimer's disease (AD) pathogenesis, there is a concern about the effect of nano-Al on cognitive function and brain health. This study was aimed to assess the effect of a 5-day oral gavage of aluminum oxide nanoparticle (nano-Al) on memory and the phosphorylation levels of hippocampal p38, JNK (c-Jun N-terminal kinase), ERK (extracellular signal-regulated kinase) as well as cleaved caspase-3 in mice. Adult male NMRI mice were treated with nano-Al in doses 5 and 10 mg/kg/oral gavage for 5 days. The test session of novel object recognition (NOR) task was performed on day 5. Following the NOR test, the hippocampi were isolated for western blot analysis to determine the total and phosphorylated levels of p38, JNK, ERK as well as cleaved caspase-3 proteins. The results showed that nano-Al oral gavage in doses of 5 and 10 mg/kg impairs NOR memory in mice. Moreover, the memory impairing effect of nano-Al coincided with a dose dependent increase in phosphorylated p38 and cleaved caspase-3 in the hippocampus. It also increased the ratio of phosphorylated to total content of ERK in the hippocampus while JNK signaling was not affected by nano-Al. This study showed that nano-Al in doses as low as 5 and 10 mg/ kg ingested for 5 days impairs NOR memory and activates p38, ERK and cleaved caspase-3 in the hippocampus.
Collapse
Affiliation(s)
- Nahid Mehrbeheshti
- Shiraz Neuroscience Research Centre, Shiraz University of Medical sciences, Shiraz, Iran
| | - Zahra Esmaili
- Nanobiology and Nanomedicine Research Centre, Shiraz University of Medical sciences, Shiraz, Iran
| | - Mojdeh Ahmadi
- Nanobiology and Nanomedicine Research Centre, Shiraz University of Medical sciences, Shiraz, Iran
| | - Maryam Moosavi
- Nanobiology and Nanomedicine Research Centre, Shiraz University of Medical sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Jiang S, Wang C, Zhu J, Huang X. Regulation of glial cell-derived neurotrophic factor in sevoflurane-induced neuronal apoptosis by long non-coding RNA CDKN2B-AS1 as a ceRNA to adsorb miR-133. Am J Transl Res 2021; 13:4760-4770. [PMID: 34150056 PMCID: PMC8205689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/16/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To investigate the regulatory mechanism of sevoflurane-induced neuronal apoptosis through analyzing the expression of glial cell-derived neurotrophic factor (GDNF) mediated by miR-133, sponged by long non-coding RNA (lncRNA) CDKN2B-AS1. METHODS An in vitro cell injury model was established by using different concentrations of sevoflurane and primary hippocampal neurons. Cell proliferation was detected by Cell Counting Kit-8 (CCK-8); caspase-3 and caspase-9 activities were detected by colorimetry, and apoptotic cells were determined by Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Fluorescence in situ hybridization (FISH) analysis was used to detect localized expression of CDKN2B antisense RNA 1 (CDKN2B-AS1), and dual-luciferase reporter assay was employed to verify the correlation of CDKN2B-AS1 and miR-133, and of miR-133 and GDNF. The expression of CDKN2B-AS1, miR-133, and GDNF mRNA in the cell injury model were measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Western blot was utilized to detect the expression of GDNF protein in the cell injury model. RESULTS In the cell injury model, CDKN2B-AS1 was highly expressed in the cytoplasm, and CDKN2B-AS1 and GDNF were downregulated and miR-133 was upregulated as detected by qRT-PCR (all P<0.05). The connections between CDKN2B-AS1 and miR-133, and between miR-133 and GDNF were confirmed. That is, CDKN2B-AS1 regulated the expression of GDNF by adsorbing miR-133 (all P<0.05). In cells treated with sevoflurane, overexpression of CDKN2B-AS1 could inhibit caspase-3 and caspase-9 activities and the degree of apoptosis. miR-133 could partially alleviate the effect of overexpressing CDKN2B-AS1 on cells, and si-GDNF the effect of miR-133 inhibitor (all P<0.05). CONCLUSION lncRNA CDKN2B-AS1 can up-regulate the expression of GDNF, inhibit neuronal apoptosis, and ease the toxic effect of sevoflurane on neural cells by acting as a sponge to adsorb miR-133.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Anesthesiology, Yanbian University Hospital (Yanbian Hospital) Yanji, Jilin Province, China
| | - Chonghe Wang
- Department of Anesthesiology, Yanbian University Hospital (Yanbian Hospital) Yanji, Jilin Province, China
| | - Jingyao Zhu
- Department of Anesthesiology, Yanbian University Hospital (Yanbian Hospital) Yanji, Jilin Province, China
| | - Xuezhu Huang
- Department of Anesthesiology, Yanbian University Hospital (Yanbian Hospital) Yanji, Jilin Province, China
| |
Collapse
|
10
|
Nam Y, Moon GJ, Kim SR. Therapeutic Potential of AAV1-Rheb(S16H) Transduction against Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22063064. [PMID: 33802760 PMCID: PMC8002454 DOI: 10.3390/ijms22063064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/08/2021] [Accepted: 03/16/2021] [Indexed: 01/19/2023] Open
Abstract
Neurotrophic factors (NTFs) are essential for cell growth, survival, synaptic plasticity, and maintenance of specific neuronal population in the central nervous system. Multiple studies have demonstrated that alterations in the levels and activities of NTFs are related to the pathology and symptoms of neurodegenerative disorders, such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and Huntington’s disease. Hence, the key molecule that can regulate the expression of NTFs is an important target for gene therapy coupling adeno-associated virus vector (AAV) gene. We have previously reported that the Ras homolog protein enriched in brain (Rheb)–mammalian target of rapamycin complex 1 (mTORC1) axis plays a vital role in preventing neuronal death in the brain of AD and PD patients. AAV transduction using a constitutively active form of Rheb exerts a neuroprotective effect through the upregulation of NTFs, thereby promoting the neurotrophic interaction between astrocytes and neurons in AD conditions. These findings suggest the role of Rheb as an important regulator of the regulatory system of NTFs to treat neurodegenerative diseases. In this review, we present an overview of the role of Rheb in neurodegenerative diseases and summarize the therapeutic potential of AAV serotype 1 (AAV1)-Rheb(S16H) transduction in the treatment of neurodegenerative disorders, focusing on diseases, such as AD and PD.
Collapse
Affiliation(s)
- Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea;
| | - Gyeong Joon Moon
- Center for Cell Therapy, Asan Institute for Life Science, Asan Medical Center, Seoul 05505, Korea;
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sang Ryong Kim
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Korea;
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- Correspondence: ; Tel.: +82-53-950-7362; Fax: +82-53-943-2762
| |
Collapse
|