1
|
Torkamani-Dordshaikh S, Darabi S, Norouzian M, Bahar R, Beirami A, Moghaddam MH, Fathi M, Vakili K, Tahmasebinia F, Bahrami M, Abbaszadeh HA, Aliaghaei A. Exploring the therapeutic potential: Apelin-13's neuroprotective effects foster sustained functional motor recovery in a rat model of Huntington's disease. Anat Cell Biol 2024; 57:419-430. [PMID: 39079710 PMCID: PMC11424562 DOI: 10.5115/acb.23.284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 08/06/2024] Open
Abstract
Huntington's disease (HD) is a hereditary condition considered by the progressive degeneration of nerve cells in the brain, resultant in motor dysfunction and cognitive impairment. Despite current treatment modalities including pharmaceuticals and various therapies, a definitive cure remains elusive. Therefore, this study investigates the therapeutic potential effect of Apelin-13 in HD management. Thirty male Wistar rats were allocated into three groups: a control group, a group with HD, and a group with both HD and administered Apelin-13. Apelin-13 was administered continuously over a 28-day period at a dosage of around 30 mg/kg to mitigate inflammation in rats subjected to 3-NP injection within an experimental HD model. Behavioral tests, such as rotarod, electromyography (EMG), elevated plus maze, and open field assessments, demonstrated that Apelin-13 improved motor function and coordination in rats injected with 3-NP. Apelin-13 treatment significantly increased neuronal density and decreased glial cell counts compared to the control group. Immunohistochemistry analysis revealed reduced gliosis and expression of inflammatory factors in the treatment group. Moreover, Apelin-13 administration led to elevated levels of glutathione and reduced reactive oxygen species (ROS) level in the treated group. Apelin-13 demonstrates neuroprotective effects, leading to improved movement and reduced inflammatory and fibrotic factors in the HD model.
Collapse
Affiliation(s)
- Shaysteh Torkamani-Dordshaikh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Norouzian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Bahar
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirreza Beirami
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Foozhan Tahmasebinia
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Bahrami
- Rayan Stem Cells and Regenerative Medicine Research Center, Ravan Sazeh Company, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Rayan Stem Cells and Regenerative Medicine Research Center, Ravan Sazeh Company, Tehran, Iran
| | - Abbas Aliaghaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Zhu D, Zhang S, Wang X, Xiao C, Cui G, Yang X. Secretory Clusterin Inhibits Dopamine Neuron Apoptosis in MPTP Mice by Preserving Autophagy Activity. Neuroscience 2024; 540:38-47. [PMID: 38242280 DOI: 10.1016/j.neuroscience.2024.01.010] [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: 03/01/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Secretory clusterin (sCLU) plays an important role in the research progress of nervous system diseases. However, the physiological function of sCLU in Parkinson's disease (PD) are unclear. The purpose of this study was to examine the effects of sCLU-mediated autophagy on cell survival and apoptosis inhibition in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. We found that MPTP administration induced prolonged pole-climbing time, shortened traction time and rotarod time, significantly decreased TH protein expression in the SN tissue of mice. In contrast, sCLU -treated mice took less time to climb the pole and had an extended traction time and rotating rod time. Meanwhile, sCLU intervention induced increased expression of the TH protein in the SN of mice. These results indicated that sCLU intervention could reduce the loss of dopamine neurons in the SN area and alleviate dyskinesia in mice. Furthermore, MPTP led to suppressed viability, enhanced apoptosis, an increased Bax/Bcl-2 ratio, and cleaved caspase-3 in the SN of mice, and these effects were abrogated by sCLU intervention. In addition, MPTP increased the levels of P62 protein, decreased Beclin1 protein, decreased the ratio of LC3B-II/LC3B-I, and decreased the numbers of autophagosomes and autophagolysosomes in the SN tissues of mice. These effects were also abrogated by sCLU intervention. Activation of PI3K/AKT/mTOR signaling with MPTP inhibited autophagy in the SN of MPTP mice; however, sCLU treatment activated autophagy in MPTP-induced PD mice by inhibiting PI3K/AKT/mTOR signaling. These data indicated that sCLU treatment had a neuroprotective effect in an MPTP-induced model of PD.
Collapse
Affiliation(s)
- Dongxue Zhu
- Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Shenyang Zhang
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaoying Wang
- Department of Ultrasound, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Chenghua Xiao
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Guiyun Cui
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xinxin Yang
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| |
Collapse
|
3
|
Tabatabaee F, Darabi S, Soltani R, Aghajanpour F, Afshar A, Abbaszadeh HA, Rajabi-Maham H. Therapeutic Effects of Exosome Therapy and Photobiomodulation Therapy on the Spermatogenesis Arrest in Male Mice After Scrotum Hyperthermia. J Lasers Med Sci 2024; 15:e3. [PMID: 38655046 PMCID: PMC11033855 DOI: 10.34172/jlms.2024.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/25/2023] [Indexed: 04/26/2024]
Abstract
Introduction: In men, several factors cause infertility, among which we can mention damage to sperm due to high temperature. So far, various treatments have been proposed for it, but they have not been highly effective. The current study aimed to evaluate the effect of exosome therapy (EXO) and photobiomodulation therapy (PBMT) on spermatogenesis arrest in male mice after scrotum hyperthermia. Methods: In this experimental study, the animals were divided into four groups: control, scrotal hyperthermia, scrotal hyperthermia+EXO (100 μL/d) (mice were treated for 30 days), scrotal hyperthermia+PBMT (laser of 0.03 J/cm2 for 30 seconds/for 30 days). Hyperthermia was induced by exposure to the temperature of 43 °C for 20 minute every day for 5 times. After 6 weeks, the animals were sacrificed. Results: The treated groups showed a significant increase in sperm parameters, as compared to the hyperthermic groups. Moreover, these favorable effects were observed in relation to the volume of testicular tissue, the number of germ cells, Leydig cells and Sertoli cells, and the level of testosterone. Research on antioxidants showed a significant reduction in oxidized glutathione (GSSG) and reactive oxygen species (ROS) in the treatment groups in comparison to the hyperthermia group (P<0.001). Also, there has been a significant increase in the amount of hydrogen peroxide enzyme observed in the hyperthermia group as opposed to the treatment group (P<0.001). Conclusion: These findings show that EXO and PBMT can improve spermatogenesis caused by hyperthermia, reduce ROS and GSSG, and increase glutathione (GSH) and sperm quality.
Collapse
Affiliation(s)
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Reza Soltani
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fakhroddin Aghajanpour
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azar Afshar
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Rajabi-Maham
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
4
|
Kirchenwitz M, Stahnke S, Grunau K, Melcher L, van Ham M, Rottner K, Steffen A, Stradal TEB. The autophagy inducer SMER28 attenuates microtubule dynamics mediating neuroprotection. Sci Rep 2022; 12:17805. [PMID: 36284196 PMCID: PMC9596692 DOI: 10.1038/s41598-022-20563-3] [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: 02/15/2022] [Accepted: 09/15/2022] [Indexed: 01/19/2023] Open
Abstract
SMER28 originated from a screen for small molecules that act as modulators of autophagy. SMER28 enhanced the clearance of autophagic substrates such as mutant huntingtin, which was additive to rapamycin-induced autophagy. Thus, SMER28 was established as a positive regulator of autophagy acting independently of the mTOR pathway, increasing autophagosome biosynthesis and attenuating mutant huntingtin-fragment toxicity in cellular- and fruit fly disease models, suggesting therapeutic potential. Despite many previous studies, molecular mechanisms mediating SMER28 activities and its direct targets have remained elusive. Here we analyzed the effects of SMER28 on cells and found that aside from autophagy induction, it significantly stabilizes microtubules and decelerates microtubule dynamics. Moreover, we report that SMER28 displays neurotrophic and neuroprotective effects at the cellular level by inducing neurite outgrowth and protecting from excitotoxin-induced axon degeneration. Finally, we compare the effects of SMER28 with other autophagy-inducing or microtubule-stabilizing drugs: whereas SMER28 and rapamycin both induce autophagy, the latter does not stabilize microtubules, and whereas both SMER28 and epothilone B stabilize microtubules, epothilone B does not stimulate autophagy. Thus, the effect of SMER28 on cells in general and neurons in particular is based on its unique spectrum of bioactivities distinct from other known microtubule-stabilizing or autophagy-inducing drugs.
Collapse
Affiliation(s)
- Marco Kirchenwitz
- grid.7490.a0000 0001 2238 295XDepartment of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany ,grid.6738.a0000 0001 1090 0254Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Stephanie Stahnke
- grid.7490.a0000 0001 2238 295XDepartment of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Kyra Grunau
- grid.7490.a0000 0001 2238 295XDepartment of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany ,grid.6738.a0000 0001 1090 0254Division of Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Lars Melcher
- grid.7490.a0000 0001 2238 295XDepartment of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Marco van Ham
- grid.7490.a0000 0001 2238 295XCellular Proteome Research, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Klemens Rottner
- grid.7490.a0000 0001 2238 295XDepartment of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany ,grid.6738.a0000 0001 1090 0254Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Anika Steffen
- grid.7490.a0000 0001 2238 295XDepartment of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Theresia E. B. Stradal
- grid.7490.a0000 0001 2238 295XDepartment of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
5
|
Gao Y, Wang C, Jiang D, An G, Jin F, Zhang J, Han G, Cui C, Jiang P. New insights into the interplay between autophagy and oxidative and endoplasmic reticulum stress in neuronal cell death and survival. Front Cell Dev Biol 2022; 10:994037. [PMID: 36187470 PMCID: PMC9524158 DOI: 10.3389/fcell.2022.994037] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Autophagy is a dynamic process that maintains the normal homeostasis of cells by digesting and degrading aging proteins and damaged organelles. The effect of autophagy on neural tissue is still a matter of debate. Some authors suggest that autophagy has a protective effect on nerve cells, whereas others suggest that autophagy also induces the death of nerve cells and aggravates nerve injury. In mammals, oxidative stress, autophagy and endoplasmic reticulum stress (ERS) constitute important defense mechanisms to help cells adapt to and survive the stress conditions caused by physiological and pathological stimuli. Under many pathophysiological conditions, oxidative stress, autophagy and ERS are integrated and amplified in cells to promote the progress of diseases. Over the past few decades, oxidative stress, autophagy and ERS and their interactions have been a hot topic in biomedical research. In this review, we summarize recent advances in understanding the interactions between oxidative stress, autophagy and ERS in neuronal cell death and survival.
Collapse
Affiliation(s)
- Yahao Gao
- Clinical Medical School, Jining Medical University, Jining, China
| | - Changshui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Di Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gang An
- Clinical Medical School, Jining Medical University, Jining, China
| | - Feng Jin
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Junchen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guangkui Han
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| | - Pei Jiang
- Department of Clinical Pharmacy, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| |
Collapse
|
6
|
Ahrabi B, Omidvari S, Mollazadeh Ghomi S, Ahmady Roozbahany N, Vafaei-Nezhad S, Shirazi Tehrani A, Abbaszadeh HA, Darabi S. Therapeutic Effects of Combination Therapy and Photobiomodulation Therapy on Retinal Regeneration. J Lasers Med Sci 2022; 13:e36. [PMID: 36743147 PMCID: PMC9841384 DOI: 10.34172/jlms.2022.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/27/2022] [Indexed: 01/27/2023]
Abstract
Introduction: Many systemic and ocular diseases cause macular edema (ME). Macular edema is seen in two primary forms; the first is diffuse thickening of the macula, and the other is a macula with a distinct petaloid (cloverleaf) appearance called cystoid macular edema. Macular edema has a known role in the reduction of visual equity, and many options have been proposed for the reversal of this condition. Methods: Articles on the effects of macular laser grid photocoagulation on diabetic macular edema (DME) or cystoid macular edema published between 2000 and 2022 were collected from PubMed, Google Scholar, and Web of Science. The following keywords were used for the search: "macular laser photocoagulation", "macular edema", "cystoid macular edema", "intravitreal pharmacotherapies", and "antivascular endothelial growth factor". Two hundred nineteen articles were found in google scholar and 165 articles in PubMed, and a total of 58 articles were included in the study after applying the exclusion criteria. Results: We investigated the effects of various lasers photocoagulation such as Focal and/or grid macular laser, subthreshold micropulse laser (SMPL), as well as intravitreal pharmacotherapies with triamcinolone acetonide, and fluocinolone, and extended released intraocular implants such as Ozurdex, Retisert, Iluvien, and anti-vascular endothelial growth factors such as bevacizumab (Avastin), Eyela, and Lucentis. Corticosteroids were more effective than lasers, although some researchers have found that lasers and combined lasers and corticosteroids are more effective. In addition, some studies have shown that the frequency and concentrations of intravitreal pharmacotherapies are effective in increasing visual outcomes. Conclusion: The results of the studies showed that the combined intravitreal corticosteroids are much more effective in improving visual acuity (VA) than a single corticosteroid, and the low concentration of the drug is safer. Still, corticosteroids have side effects such as increased intraocular pressure and glaucoma. Therefore, combining the medication with a laser is much more reasonable than each alone. Also, the subthreshold photocoagulation laser (670 nm) is better at reducing the central macular thickness (CMT) and improving VA than the micro pulse yellow laser and pan-retinal photocoagulation (PRP).
Collapse
Affiliation(s)
- Behnaz Ahrabi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samareh Omidvari
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shamim Mollazadeh Ghomi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Navid Ahmady Roozbahany
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Vafaei-Nezhad
- Department of Anatomical Sciences, School of Medicine, Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Atefeh Shirazi Tehrani
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Correspondence to Hojjat Allah Abbaszadeh, Shahram Darabi,
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran,Correspondence to Hojjat Allah Abbaszadeh, Shahram Darabi,
| |
Collapse
|
7
|
Ahrabi B, Abbaszadeh HA, Piryaei A, Shekari F, Ahmady Roozbahany N, Rouhollahi M, Azam Sayahpour F, Ahrabi M, Azimi H, Moghadasali R. Autophagy-induced mesenchymal stem cell-derived extracellular vesicles ameliorated renal fibrosis in an in vitro model. BIOIMPACTS : BI 2022; 13:359-372. [PMID: 37736337 PMCID: PMC10509741 DOI: 10.34172/bi.2022.24256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 09/23/2023]
Abstract
Introduction Chronic and progressive damage to the kidney by inflammatory processes, may lead to an increase in the extracellular matrix production, a condition known as renal fibrosis. The current study aims to evaluate if the extracellular vesicles (EVs) derived from autophagic adipose-derived mesenchymal stem cells (ADMSCs) can reduce the inflammation and extracellular matrix accumulation in damaged kidney tissue. Methods Autophagy was induced in ADMSCs using 2µM concentration curcumin and was confirmed by evaluating LC3B, ATG7, and Beclin1 using real-time polymerase chain reaction (PCR) and Western blot. An in vitro renal fibrotic model was established in HEK-293 cells exposed to H2O2 (0.8mM) for 24 and 72 hours. The fibrotic model was confirmed through evaluation of collagen I, transforming growth factor-beta 1 (TGF-β1), E-cadherin, and vimentin genes expression using real-time PCR, collagen I protein by ELISA. After induction of fibrosis for 24 and 72 hours, the HEK cells were treated with NEVs (non-autophagy EVs) (50µM) or AEVs (autophagy EVs) (50µM) at 48, 96, and 124 hours, and then the samples were collected at 72 and 148 hours. Expression of collagen I, TGF-β1, E-cadherin, and vimentin Genes was evaluated via RT-PCR, and protein levels of IL1, TNF-α, IL4, IL10 using ELISA. Results Induction of autophagy using curcumin (2µM) for 24 hours significantly increased LC3B, Beclin1, and ATG7 in the ADMSCs. Upregulation in anti-fibrotic (E-cadherin) and anti-inflammatory (IL4, IL10) gene expression was significantly different in the fibrotic model treated by AEVs compared to NEVs. Also, the downregulation of fibrotic (TGF-β1, vimentin, collagen I) and pro-inflammatory (IL1, TNFα) gene expression was significantly different in AEVs compared with those treated by NEVs. Conclusion Our findings suggest that AEVs can be considered as a therapeutic modality for renal fibrosis in the future.
Collapse
Affiliation(s)
- Behnaz Ahrabi
- Department of Biology and Anatomical Sciences, school of medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Department of Biology and Anatomical Sciences, school of medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, school of medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | | | - Mahya Rouhollahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahnaz Ahrabi
- Department of Endodontics, Dental Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Azimi
- Department of English Language Teaching, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
8
|
Vafaei-Nezhad S, Niknazar S, Payvandi AA, Shirazi Tehrani A, Ahmady Roozbahany N, Ahrabi B, Abbaszadeh HA, Darabi S. Therapeutic Effects of Photobiomodulation Therapy on Multiple Sclerosis by Regulating the Inflammatory Process and Controlling Immune Cell Activity: A Novel Promising Treatment Target. J Lasers Med Sci 2022; 13:e32. [PMID: 36743142 PMCID: PMC9841388 DOI: 10.34172/jlms.2022.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022]
Abstract
Introduction: Multiple sclerosis (MS) is one of the autoimmune and chronic diseases of the central nervous system; this disease occurs more frequently in young people and women and leads to neurological symptoms. Oxidative stress, inflammatory processes, and oligodendrocyte dysfunction have a pivotal role in the pathophysiology of this disease. Nowadays it is reported that photobiomodulation (PBM) as a non-invasive treatment has neuroprotective potential, but the exact mechanisms are not understood. Methods: In this study, we reviewed the effects of PBM on MS. In this regard, we used the keywords "Photobiomodulation", "Laser therapy", and "Low-level laser therapy" on MS to find related studies on this subject in PubMed, Google scholar, Elsevier, Medline, and Scopus databases. Results: PBM has positive effects on MS by regulating the inflammatory process, controlling immune cell activity and mitochondrial functions, as well as inhibiting free radicals production which finally leads to a reduction in neurological defects and an improvement in the functional status of patients. Conclusion: Overall, researchers have suggested the use of laser therapy in neurodegenerative diseases due to its numerous therapeutic effects.
Collapse
Affiliation(s)
- Saeed Vafaei-Nezhad
- Department of Anatomical Sciences, School of Medicine, Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran,Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Niknazar
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Asghar Payvandi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefeh Shirazi Tehrani
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Ahmady Roozbahany
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnaz Ahrabi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Correspondence to Hojjat Allah Abbaszadeh, Hearing Disorders Research Center, Loghman Hakim Hospital;
; Shahram Darabi, Cellular and Molecular Research Center, Research Institute for NonCommunicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran;
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran,Correspondence to Hojjat Allah Abbaszadeh, Hearing Disorders Research Center, Loghman Hakim Hospital;
; Shahram Darabi, Cellular and Molecular Research Center, Research Institute for NonCommunicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran;
| |
Collapse
|
9
|
Compounds activating VCP D1 ATPase enhance both autophagic and proteasomal neurotoxic protein clearance. Nat Commun 2022; 13:4146. [PMID: 35842429 PMCID: PMC9288506 DOI: 10.1038/s41467-022-31905-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 07/08/2022] [Indexed: 01/04/2023] Open
Abstract
Enhancing the removal of aggregate-prone toxic proteins is a rational therapeutic strategy for a number of neurodegenerative diseases, especially Huntington's disease and various spinocerebellar ataxias. Ideally, such approaches should preferentially clear the mutant/misfolded species, while having minimal impact on the stability of wild-type/normally-folded proteins. Furthermore, activation of both ubiquitin-proteasome and autophagy-lysosome routes may be advantageous, as this would allow effective clearance of both monomeric and oligomeric species, the latter which are inaccessible to the proteasome. Here we find that compounds that activate the D1 ATPase activity of VCP/p97 fulfill these requirements. Such effects are seen with small molecule VCP activators like SMER28, which activate autophagosome biogenesis by enhancing interactions of PI3K complex components to increase PI(3)P production, and also accelerate VCP-dependent proteasomal clearance of such substrates. Thus, this mode of VCP activation may be a very attractive target for many neurodegenerative diseases.
Collapse
|
10
|
Ahrabi B, Tabatabaei Mirakabad FS, Niknazar S, Payvandi AA, Ahmady Roozbahany N, Ahrabi M, Torkamani SD, Abbaszadeh HA. Photobiomodulation Therapy and Cell Therapy Improved Parkinson's Diseases by Neuro-regeneration and Tremor Inhibition. J Lasers Med Sci 2022; 13:e28. [PMID: 36743130 PMCID: PMC9841383 DOI: 10.34172/jlms.2022.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/02/2022] [Indexed: 11/22/2022]
Abstract
Introduction: Parkinson's disease (PD) is a progressive and severe neurodegenerative disorder of the central nervous system (CNS). The most prominent features of this disease are cell reduction in the substantia nigra and accumulation of α-synuclein, especially in the brainstem, spinal cord, and cortical areas. In addition to drug-based treatment, other therapies such as surgery, cell therapy, and laser therapy can be considered. In this study, articles on cell therapy and laser therapy for PD have been collected to evaluate the improvement of motor function, cell differentiation, and dopaminergic cell proliferation. Methods: Articles were collected from four electronic databases: PubMed, Scopus, Google Scholar, and Web of Science from 2010 to 2022. The keywords were "photobiomodulation", "low-level light therapy", "Low-level laser therapy", "near-infrared light", "Parkinson's disease", "Parkinsonism", and "stem cell therapy". About 100 related articles were included in the study. Results: The results of the studies showed that cell therapy and laser therapy are useful in the treatment of PD, and despite their limitations, they can be useful in improving PD. Conclusion: Concomitant use of cell therapy and photobiomodulation therapy can improve the symptoms of PD.
Collapse
Affiliation(s)
- Behnaz Ahrabi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Somayeh Niknazar
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Asghar Payvandi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mahnaz Ahrabi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaysteh Dordshaikh Torkamani
- Department of Anatomical Sciences and Biology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Department of Anatomical Sciences and Biology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Correspondence to Hojjat-Allah Abbaszadeh, Laser Application in Medical Sciences Research Center and Department of Biology and Anatomical Sciences, school of medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. P.O. Box: 19395-4719. Tel: +98-21-23872555;
| |
Collapse
|
11
|
Ebrahimi-Kia Y, Noori-Zadeh A, Rajaei F, Darabi S, Darabi L, Ghasemi Hamidabadi H. The Effect of bisphenol A and Photobiomodulation Therapy on Autophagy-Related Genes Induction in Adipose Tissue-Derived Stem Cells. J Lasers Med Sci 2022; 13:e15. [DOI: 10.34172/jlms.2022.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 12/18/2021] [Indexed: 11/09/2022]
Abstract
Introduction: As adipose tissue-derived stem cells (ADSCs) can divide rapidly and be prepared non-invasively, they have extensively been used in regenerative medicine. On the other hand, a new method of therapy, known as photobiomodulation (PHT), has been used to treat many diseases, such as inflammatory conditions, wound healing and pain. Besides, exposure to chemical substances such as bisphenol A (BPA), at low levels, can lead to autophagy. This study investigated the effects of BPA and PHT on the expression of autophagy-related genes, including LC3, NRF2, P62, in rat ADSCs as a model. Methods: ADSCs isolation and purification were confirmed by immunocytochemistry (ICC). The cells were then treated with different concentrations of BPA and also subjected to PHT. Reverse transcription polymerase chain reaction (RT-PCR) was used for the evaluation of LC3, NRF2 and P62 gene expressions. Oil red O staining was used for adipogenic vacuole formation. Result: ICC showed that the isolated cells were CD 49-positive but CD 31 and CD 34-negative. The viability test indicated that the number of live cells after 24 hours in the BPA groups at concentrations of 0, 1, 50, 100 and 200 μM was 100%, 93%, 81%, 72%, and 43% respectively. The difference in cell viability between groups 50, 100 and 200 μM was significant as compared with the control groups (P<0.05). Moreover, in the group with 1 μM concentration of BPA, the expressions of LC3, NRF2 and P62 genes were upregulated. However, in the treatment group at the concentration of 200 μM of BPA, the LC3 gene was expressed, but NRF2 and P62 genes were downregulated. Conclusion: BPA and PHT induce autophagy and adiposeness in ADSCs in a dose-dependent manner.
Collapse
Affiliation(s)
| | - Ali Noori-Zadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
- Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Leila Darabi
- Department of Neurology, Islamic Azad University Tehran Medical branch (IAUTMU), Tehran, Iran
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
12
|
Tehrani AS, Mirakabad FST, Abdollahifar MA, Mollazadehghomi S, Darabi S, Forozesh M, Rezaei-Tavirani M, Mahmoudiasl GR, Ahrabi B, Azimzadeh Z, Abbaszadeh HA. Severe Acute Respiratory Syndrome Coronavirus 2 Induces Hepatocyte Cell Death, Active Autophagosome Formation and Caspase 3 Up-Regulation in Postmortem Cases: Stereological and Molecular Study. TOHOKU J EXP MED 2022; 256:309-319. [PMID: 35321977 DOI: 10.1620/tjem.2022.j007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Atefeh Shirazi Tehrani
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences.,Hearing disorders research center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences
| | | | - Mohammad-Amin Abdollahifar
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences.,Hearing disorders research center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences
| | | | - Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences
| | | | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Para medicine, Shahid Beheshti University of Medical Sciences
| | | | - Behnaz Ahrabi
- Department of Biology and Anatomy, Shahid Beheshti University of Medical Sciences
| | - Zahra Azimzadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences.,Hearing disorders research center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences.,Department of Biology and Anatomy, Shahid Beheshti University of Medical Sciences
| |
Collapse
|
13
|
Wang Y, Gao L, Chen J, Li Q, Huo L, Wang Y, Wang H, Du J. Pharmacological Modulation of Nrf2/HO-1 Signaling Pathway as a Therapeutic Target of Parkinson's Disease. Front Pharmacol 2021; 12:757161. [PMID: 34887759 PMCID: PMC8650509 DOI: 10.3389/fphar.2021.757161] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder featuring both motor and nonmotor symptoms associated with a progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Oxidative stress (OS) has been implicated in the pathogenesis of PD. Genetic and environmental factors can produce OS, which has been implicated as a core contributor to the initiation and progression of PD through the degeneration of dopaminergic neurons. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) orchestrates activation of multiple protective genes, including heme oxygenase-1 (HO-1), which protects cells from OS. Nrf2 has also been shown to exert anti-inflammatory effects and modulate both mitochondrial function and biogenesis. Recently, a series of studies have reported that different bioactive compounds were shown to be able to activate Nrf2/antioxidant response element (ARE) and can ameliorate PD-associated neurotoxin, both in animal models and in tissue culture. In this review, we briefly overview the sources of OS and the association between OS and the pathogenesis of PD. Then, we provided a concise overview of Nrf2/ARE pathway and delineated the role played by activation of Nrf2/HO-1 in PD. At last, we expand our discussion to the neuroprotective effects of pharmacological modulation of Nrf2/HO-1 by bioactive compounds and the potential application of Nrf2 activators for the treatment of PD. This review suggests that pharmacological modulation of Nrf2/HO-1 signaling pathway by bioactive compounds is a therapeutic target of PD.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Luyan Gao
- Department of Neurology, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Central Clinical College, Tianjin Medical University, Tianjin, China
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Qiang Li
- Department of Neurology, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Liang Huo
- Department of Pediatric Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanchao Wang
- Department of Neurology, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Hongquan Wang
- Department of Neurology, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Jichen Du
- Department of Neurology, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| |
Collapse
|
14
|
Deng H, Ma Z. Protective effects of berberine against MPTP-induced dopaminergic neuron injury through promoting autophagy in mice. Food Funct 2021; 12:8366-8375. [PMID: 34342315 DOI: 10.1039/d1fo01360b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Berberine, an isoquinoline alkaloid isolated from Coptis chinensis, has been widely studied for its efficacy in the treatment of neurodegenerative diseases. However, the detailed mechanisms are unknown. In this study, the effects of berberine on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice model of Parkinson's disease were investigated. We showed that treatment with berberine significantly ameliorates the degeneration of dopaminergic neurons in substantia nigra compacta (SNc) and improves motor impairment in MPTP-treated mice. Berberine also significantly decreased the level of α-synuclein and enhanced the microtubule-associated protein light chain 3 (LC3-II)-associated autophagy in the SN of MPTP-treated mice. Furthermore, adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) was activated by berberine. Berberine's actions were abolished by pre-treatment with 3-methyladenine (an autophagy inhibitor) or compound c (an AMPK inhibitor) in the MPP+-treated SH-SY5Y cells. These results suggested that the protective effects of berberine on the toxicity of MPTP could be attributed to berberine-enhanced autophagy via the AMPK dependent pathway.
Collapse
Affiliation(s)
- Han Deng
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China.
| | - ZeGang Ma
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, China. and Institute of Brain Science and Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
15
|
A bibenzyl compound 20C protects rats against 6-OHDA-induced damage by regulating adaptive immunity associated molecules. Int Immunopharmacol 2020; 91:107269. [PMID: 33340781 DOI: 10.1016/j.intimp.2020.107269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/22/2020] [Accepted: 11/29/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with complicated pathogenesis. A novel bibenzyl compound 2-[4-hydroxy-3-(4-hydroxyphenyl)benzyl]-4-(4-hydroxyphenyl)phenol (20C) has been shown to have some neuroprotective effects, and its mechanism still needs further research. In this study, we used a 6-hydroxydopamine (6-OHDA)-induced PD rat model to evaluate the protective effect of 20C. Our study found that 20C could improve behavioral defects in 6-OHDA-lesion rats, decrease neuroinflammation and protect their DA neurons. It could inhibit the activity of inducible nitric oxide synthase (iNOS) induced by 6-OHDA, and lead to a decrease in the expression of nitrated-α-synuclein. When exposed to AMT-an inhibitor of iNOS, the nitrated-α-synuclein in PC12 decreased, and 20C demonstrated the same function on nitrated-α-synuclein as AMT. Besides, we also found that nitrated-α-synuclein was displayed in microglia. And 20C could decrease the expression of antigen-presenting molecule major histocompatibility complex I (MHC I) in dopamine (DA) neurons and MHC II in microglia induced by 6-OHDA. So, these imply that nitrated-α-synuclein might act as an endogenous antigen activating adaptive immunity, and the neuroprotection of 20C might be associated with inhibiting the activity of iNOS, decreasing the expression of the antigen molecule nitrated-α-synuclein and the antigen presenting molecule MHC. Our results indicated that inhibiting iNOS might be an effective strategy to protect neurons from oxidative stress.
Collapse
|
16
|
Ghrelin protects dopaminergic neurons against MPTP neurotoxicity through promoting autophagy and inhibiting endoplasmic reticulum mediated apoptosis. Brain Res 2020; 1746:147023. [PMID: 32710901 DOI: 10.1016/j.brainres.2020.147023] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder, the important pathology of PD due to the prominent loss of the dopaminergic neurodegeneration in the substantia nigra pars compacta (SNpc) and striatum (STR). Although the etiology of PD is not fully understood, aggregation of α-synuclein, impaired autophagy, and endoplasmic reticulum stress (ERS) are involved in the pathogenesis of PD. Previously it has been demonstrated that Ghrelin is a kind of peptide protected dopaminergic neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyran (MPTP)-induced neurotoxicity, but the detailed mechanism remains to be elucidated. In the present work, we investigated the effects of Ghrelin on autophagy and ERS-mediated apoptosis in the MPTP-lesioned PD mice model. We found that Ghrelin was neuroprotective against MPTP-induced dopaminergic neurodegeneration. Subsequently, we investigated Ghrelin inhibited the accumulation and phosphorylation of α-synuclein induced by MPTP. Moreover, Ghrelin promoted autophagy indicated by the up-regulation of microtubule-associated protein 1 Light Chain 3B-II/I (LC3B-II/I) and Beclin1, as well as decreasing the level of p62 in the SNpc and STR. Besides, the activation of the ERS-related apoptosis signaling pathway including IRE1α and Caspase-12 signaling pathway induced by MPTP was suppressed by Ghrelin treatment. Furthermore, Ghrelin also decreased Caspase-3 expression. Taken together, our results indicated that Ghrelin may exert neuroprotective effects via regulating α-synuclein activities, enhancing autophagy, and ameliorating ERS-mediated apoptosis in MPTP-lesioned mice, which provides a new target for potential pharmacologic interventions of PD treatment in the future.
Collapse
|
17
|
Zhong X, Wang B, Zhang G, Yuan Y, Hu X, Xiong J, Zheng P, Liu Y, Xu K, Xiao J, Wu Y, Ye J. Autophagy Activation Is Involved in Acidic Fibroblast Growth Factor Ameliorating Parkinson's Disease via Regulating Tribbles Homologue 3. Front Pharmacol 2019; 10:1428. [PMID: 31849673 PMCID: PMC6901012 DOI: 10.3389/fphar.2019.01428] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a degenerative disorder of the central nervous system, resulting in loss of dopamine neurons. Excessive endoplasmic reticulum (ER) stress and autophagy dysfunction play a crucial role on Parkinson's disease (PD) development. It has been showed that acidic fibroblast growth factor (aFGF) alleviates the development of PD by inhibiting ER stress. But the role of autophagy and its relationship with ER stress during aFGF treatment for PD has not been elucidated. We found that both aFGF and rapamycin (Rapa) improved 6-Hydroxy Dopamine (6-OHDA)-induced PD development as shown with histomorphology results in striatum and substantia nigra (SNpc). Additionally, aFGF promoted autophagy with increasing mTOR and decreasing p62 expressions, and then exerts its neuroprotective role in 6-OHDA-treated PC12 cells, which were abolished by chloroquine (CQ) treatment. Moreover, 4-phenylbutyric acid (4-PBA) administration inhibited the expressions of autophagy markers during 6-OHDA-treated PC12 cells, which was similar with aFGF treating PC12 cells under 6-OHDA condition. Furthermore, we had detected the expressions of CHOP and its downstream factor, tribbles homologue 3 (TRB3), a pro-apoptotic protein. We found that TRB3 and CHOP expressions were significantly downregulated after treating with aFGF and 4-PBA in 6-OHDA-treated PC12 cells and PD model. Taken together, this study has demonstrated that aFGF treatment ameliorates 6-OHDA-induced elevated ER stress and subsequently suppression of autophagy via inhibiting TRB3 activation, and consequently ameliorates 6-OHDA-induced neurotoxicity.
Collapse
Affiliation(s)
- Xingfeng Zhong
- Department of Anesthesia, The First Affiliated Hospital, Gannan Medical University, Ganzhou, China.,Department of Anesthesia, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Beini Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Guanyinsheng Zhang
- Department of Anesthesia, The First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Yuan Yuan
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Xiaoli Hu
- Department of Anesthesia, The First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Jun Xiong
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Peipei Zheng
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yaqian Liu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Ke Xu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Junming Ye
- Department of Anesthesia, The First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| |
Collapse
|
18
|
Zhu J, Gao W, Shan X, Wang C, Wang H, Shao Z, Dou S, Jiang Y, Wang C, Cheng B. Apelin-36 mediates neuroprotective effects by regulating oxidative stress, autophagy and apoptosis in MPTP-induced Parkinson's disease model mice. Brain Res 2019; 1726:146493. [PMID: 31586624 DOI: 10.1016/j.brainres.2019.146493] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/28/2019] [Accepted: 10/02/2019] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD), a common human neurodegenerative disorder, is characterized by the presence of intraneuronal Lewy bodies composed principally of abnormal aggregated and post-translationally modified α-synuclein. In our previous research, we have demonstrated the neuroprotective effect of Apelin-36, a neuroendocrine peptide in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)-lesioned PD model mice. Therefore, this study was designed to evaluate the neuroprotective mechanism of Apelin-36 against MPTP-induced neurotoxicity in mice. The results showed that MPTP-induced the depletion of dopamine in the striatum (STR) was partially reversed by Apelin-36. Apelin-36 also improved the activity of antioxidant system including superoxide dismutase (SOD) and glutathione (GSH), and decreased the overproduction of malondialdehyde (MDA) in the substantia nigra pars compacta (SNpc) and STR of MPTP-treated mice. Moreover, Apelin-36 downregulated inducible nitric oxide synthase (iNOS) and nitrated α-synuclein expression. Furthermore, Apelin-36 significantly promoted autophagy indicated by the up-regulation of LC3-II and Beclin1 and inhibition of p62 expression in the SNpc and STR of MPTP-treated mice. The protective effect of Apelin-36 was also associated with the inhibition of the apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) signaling pathway and inactivation of caspase-3. Taken together, our findings demonstrated that the neuroprotective mechanism of Apelin-36 against MPTP-induced neurotoxicity in mice might be related to decreasing the aggregation of nitrated α-synuclein and alleviating oxidative stress as well as promoting autophagy and inhibiting ASK1/JNK/caspase-3 apoptotic pathway, which provides a novel strategy for PD treatment.
Collapse
Affiliation(s)
- Junge Zhu
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Wenming Gao
- Basic Medical Sciences, Jining Medical University, 272067 Jining, China
| | - Xuehua Shan
- Basic Medical Sciences, Jining Medical University, 272067 Jining, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Huiqing Wang
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Ziqi Shao
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Shanshan Dou
- Basic Medical Sciences, Jining Medical University, 272067 Jining, China
| | - Yunlu Jiang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Chuangong Wang
- Basic Medical Sciences, Jining Medical University, 272067 Jining, China.
| | - Baohua Cheng
- Neurobiology Institute, Jining Medical University, 272067 Jining, China.
| |
Collapse
|
19
|
Darabi S, Noori-Zadeh A, Rajaei F, Abbaszadeh HA, Abdollahifar MA, Bakhtiyari S. Neuroprotective Effects of the Small-Molecule Enhancer of Rapamycin in the Cellular Model of Parkinson’s Disease. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09798-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|