1
|
Naghshbandieh A, Naghshbandieh A, Barfi E, Abkhooie L. Assessment of the level of apoptosis in differentiated pseudo-neuronal cells derived from neural stem cells under the influence of various inducers. AMERICAN JOURNAL OF STEM CELLS 2024; 13:250-270. [PMID: 39850017 PMCID: PMC11751472 DOI: 10.62347/bptg6174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/23/2024] [Indexed: 01/25/2025]
Abstract
Development and maintenance of the nervous system are governed by a scheduled cell death mechanism known as apoptosis. Very much how neurons survive and function depends on the degree of death in differentiating pseudo-neuronal cells produced from neural stem cells. Different inducers can affect the degree of death in these cells: hormones, medicines, growth factors, and others. Developing inventive therapies for neurodegenerative illnesses depends on a knowledge of how these inducers impact mortality in differentiated pseudo-neuronal cells. Using flow cytometry, Western blotting, and fluorescence microscopy among other techniques, the degree of death in many pseudo-neuronal cells is evaluated. Flow cytometry generates dead cell counts from measurements of cell size, granularity, and DNA content. Whereas fluorescence microscopy visualizes dead cells using fluorescent dyes or antibodies, Western blotting detects caspases and Bcl-2 family proteins. This review attempts to offer a thorough investigation of present studies on death in differentiated pseudo-neuronal cells produced from neural stem cells under the effect of different inducers. Through investigating how these inducers influence death, the review aims to provide information that might direct the next studies and support treatment plans for neurodegenerative diseases. With an eye toward inducers like retinoic acid, selegiline, cytokines, valproic acid, and small compounds, we examined research to evaluate death rates. The findings offer important new perspectives on the molecular processes guiding death in these cells. There is still a complete lack of understanding of how different factors affect the molecular processes that lead to death, so understanding these processes can contribute to new therapeutic approaches to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Adele Naghshbandieh
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares UniversityTehran, Iran
| | - Atefe Naghshbandieh
- Department of Pharmaceutical Biotechnology and Department of Pharmaceutical and Bimolecular Science, University of MilanMilan, Italy
| | - Elahe Barfi
- Razi Herbal Medicines Research Center, Lorestan University of Medical SciencesKhorramabad, Iran
| | - Leila Abkhooie
- Razi Herbal Medicines Research Center, Lorestan University of Medical SciencesKhorramabad, Iran
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical SciencesKhorramabad, Iran
| |
Collapse
|
2
|
Li J, Bu Y, Li B, Zhang H, Guo J, Hu J, Zhang Y. Calenduloside E alleviates cerebral ischemia/reperfusion injury by preserving mitochondrial function. J Mol Histol 2022; 53:713-727. [PMID: 35819738 PMCID: PMC9374638 DOI: 10.1007/s10735-022-10087-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/31/2022] [Indexed: 12/16/2022]
Abstract
Calenduloside E (CE) isolated from Aralia elata (Miq.) Seem. is a natural triterpenoid saponin that can reportedly ameliorate myocardial ischemia/reperfusion injury. However, its potential roles and mechanism in cerebral ischemia/reperfusion injury are barely understood. In this study, we established an oxygen-glucose deprivation/reoxygenation (OGD/R) model in HT22 cells. We found that CE significantly attenuated the OGD/R-induced inhibition of cell viability and apoptotic cell death in HT22 cells. Moreover, CE treatment significantly ameliorated OGD/R-induced mitochondrial fission by inhibiting mitochondrial dynamin-related protein 1 (Drp1) recruitment and increasing Drp1 phosphorylation at Ser637. CE treatment significantly ameliorated OGD/R-induced mitochondrial dysfunction by increasing the mitochondrial membrane potential and reducing the mitochondrial ROS and cellular calcium accumulation. Moreover, CE treatment significantly inhibited the OGD/R-induced release of mitochondrial Cytochrome C and increase in Bax, Cleaved-caspase3 and Cleaved-caspase9 protein levels, whereas CE treatment significantly reversed the OGD/R-induced decrease in Bcl-2 and full length of caspase3 and caspase9 protein levels. In vivo, we found that CE treatment significantly ameliorated ischemic/hypoxic-induced brain infarct volume, neurological deficits, and neuronal apoptosis in mice after middle cerebral artery occlusion and reperfusion. CE treatment also significantly ameliorated the mitochondrial transmembrane potential, decreased Cytochrome C release, and reversed the increase in Bax, Cleaved-caspase3 and Cleaved-caspase9 protein levels and the decrease in Bcl-2 and full length of caspase3 and caspase9 protein levels induced by cerebral ischemia/reperfusion (I/R). All these results indicated that CE treatment exerted a neuroprotective effect by ameliorating mitochondrial dysfunction during cerebral I/R injury.
Collapse
Affiliation(s)
- Jianxiong Li
- Department of Neurology, Lanzhou University Second Hospital, 730030, Lanzhou, Gansu Province, China
| | - Yujie Bu
- Department of Neurology, Lanzhou University Second Hospital, 730030, Lanzhou, Gansu Province, China
| | - Bin Li
- Department of Neurology, Lanzhou University Second Hospital, 730030, Lanzhou, Gansu Province, China
| | - Hailin Zhang
- Neurosurgery, Lanzhou University Second Hospital, 730030, Lanzhou, Gansu Province, China.
| | - Jia Guo
- Department of Neurology, Lanzhou University Second Hospital, 730030, Lanzhou, Gansu Province, China
| | - Jianping Hu
- Department of Neurology, Lanzhou University Second Hospital, 730030, Lanzhou, Gansu Province, China
| | - Yanfang Zhang
- Department of Neurology, Lanzhou University Second Hospital, 730030, Lanzhou, Gansu Province, China
| |
Collapse
|
3
|
Hong S, Lee SE, Kang I, Yang J, Kim H, Kim J, Kang KS. Induced neural stem cells from human patient-derived fibroblasts attenuate neurodegeneration in Niemann-Pick type C mice. J Vet Sci 2021; 22:e7. [PMID: 33522159 PMCID: PMC7850792 DOI: 10.4142/jvs.2021.22.e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/25/2020] [Accepted: 10/28/2020] [Indexed: 11/20/2022] Open
Abstract
Background Niemann-Pick disease type C (NPC) is caused by the mutation of NPC genes, which leads to the abnormal accumulation of unesterified cholesterol and glycolipids in lysosomes. This autosomal recessive disease is characterized by liver dysfunction, hepatosplenomegaly, and progressive neurodegeneration. Recently, the application of induced neural stem cells (iNSCs), converted from fibroblasts using specific transcription factors, to repair degenerated lesions has been considered a novel therapy. Objectives The therapeutic effects on NPC by human iNSCs generated by our research group have not yet been studied in vivo; in this study, we investigate those effects. Methods We used an NPC mouse model to efficiently evaluate the therapeutic effect of iNSCs, because neurodegeneration progress is rapid in NPC. In addition, application of human iNSCs from NPC patient-derived fibroblasts in an NPC model in vivo can give insight into the clinical usefulness of iNSC treatment. The iNSCs, generated from NPC patient-derived fibroblasts using the SOX2 and HMGA2 reprogramming factors, were transplanted by intracerebral injection into NPC mice. Results Transplantation of iNSCs showed positive results in survival and body weight change in vivo. Additionally, iNSC-treated mice showed improved learning and memory in behavior test results. Furthermore, through magnetic resonance imaging and histopathological assessments, we observed delayed neurodegeneration in NPC mouse brains. Conclusions iNSCs converted from patient-derived fibroblasts can become another choice of treatment for neurodegenerative diseases such as NPC.
Collapse
Affiliation(s)
- Saetbyul Hong
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Seung Eun Lee
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Insung Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jehoon Yang
- Animal Research and Molecular Imaging Center, Samsung Medical Center, Seoul 06351, Korea
| | - Hunnyun Kim
- Animal Research and Molecular Imaging Center, Samsung Medical Center, Seoul 06351, Korea
| | - Jeyun Kim
- Animal Research and Molecular Imaging Center, Samsung Medical Center, Seoul 06351, Korea
| | - Kyung Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
4
|
Zhang J, Zhu M, Zhang J. Pachypodol protects newborn rats from anaesthesia-induced apoptosis in the developing brain by regulating the JNK/ERK pathway. Int J Dev Neurosci 2021; 81:633-642. [PMID: 34198359 DOI: 10.1002/jdn.10140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/11/2022] Open
Abstract
Anaesthesia exposure causes changes in the developing brain and affects behaviour and memory. This study examined the beneficial effect of pachypodol against isoflurane (ISF)-induced neuronal injury. Seven-day-old rats were treated with 10 mg/kg and 30 mg/kg intravenous pachypodol 30 min before exposure to ISF (0.75%) for 6 h. Oxidative stress and other biochemical parameters were assessed in the brain tissue and serum using enzyme-linked immunosorbent assay. Additionally, a terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) assay was performed to assess neuronal cell apoptosis in several regions of the hippocampus. Cognitive function and neurological scores were determined in the pachypodol-treated neuron-injured rats. Cytokine levels and oxidative stress were reduced in the pachypodol-treated group compared with the ISF group. In addition, cognitive deterioration was reversed in pachypodol-treated compared with ISF-treated rats. Thus, treatment with pachypodol reduced neuronal apoptosis in neuron-injured rats. Moreover, pachypodol ameliorated changes to the JNK/ERK/Akt pathway in brain-injured rats. In conclusion, pachypodol treatment prevents neuronal apoptosis in ISF-treated rats by regulating the JNK/ERK pathway.
Collapse
Affiliation(s)
- Jingxiong Zhang
- Department of Anesthesiology, Huashan Hospital North Affiliated to Fudan University, Shanghai, China
| | - Mingsheng Zhu
- Department of Anesthesiology, Huashan Hospital North Affiliated to Fudan University, Shanghai, China
| | - Jie Zhang
- Department of Anesthesiology, Huashan Hospital North Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
5
|
Liang F, Fu X, Li Y, Han F. Desoxyrhapontigenin attenuates neuronal apoptosis in an isoflurane-induced neuronal injury model by modulating the TLR-4/cyclin B1/Sirt-1 pathway. AMB Express 2020; 10:175. [PMID: 32997222 PMCID: PMC7527400 DOI: 10.1186/s13568-020-01105-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/09/2020] [Indexed: 02/08/2023] Open
Abstract
This study investigated the protective effect of desoxyrhapontigenin (DOP) against isoflurane (ISF)-induced neuronal injury in rats. Neuronal injury was induced in pups by exposing them to 0.75% ISF on postnatal day 7 with 30% oxygen for 6 h. The pups were treated with DOP 10 mg/kg, i.p., for 21 days after ISF exposure. The protective effect of DOP was estimated by assessing cognitive function using the neurological score and the Morris water maze. Neuronal apoptosis was assessed in the hippocampus using the TUNEL assay, and protein expression of caspase-3, Bax, and Bcl-2 was measured by Western blotting. The levels of cytokines and oxidative stress parameters were assessed by ELISA. Western blotting and RT-PCR were performed to measure the expression of NF-kB, TLR-4, Sirt-1, and cyclin B1 protein in the brain. The cognitive function and neurological function scores were improved in the DOP group compared with the ISF group. Moreover, DOP treatment reduced the number of TUNEL-positive cells and the expression of caspase-3, Bax, and Bcl-2 protein in the brains of rats with neuronal injury. The levels of mediators of inflammation and oxidative stress were reduced in the brain tissue of the DOP group. Treatment with DOP attenuated the protein expression of TLR-4, NF-kB, cyclin B1, and Sirt-1 in the brain tissue of rats with neuronal injury. In conclusion, DOP ameliorates neuronal apoptosis and improves cognitive function in rats with ISF-induced neuronal injury. Moreover, DOP treatment can prevent neuronal injury by regulating the TLR-4/cyclin B1/Sirt-1 pathway.
Collapse
|
6
|
Tandean S, Japardi I, Loe ML, Riawan W, July J. Protective Effects of Propolis Extract in a Rat Model of Traumatic Brain Injury via Hsp70 Induction. Open Access Maced J Med Sci 2019; 7:2763-2766. [PMID: 31844433 PMCID: PMC6901855 DOI: 10.3889/oamjms.2019.736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND: Traumatic brain injury (TBI) is one of the major global health problems. Secondary brain injury is a complex inflammation cascades process that causes brain cell apoptosis. Propolis is a natural product that has neuroprotective property. AIM: This study aimed to investigate the effect of propolis toward Hsp70 expression with apoptosis marker in brain tissue after TBI. METHODS: Thirty-three Sprague Dawley rats were randomised into three treatments group, i.e. sham-operated controls, closed head injury (CHI), and CHI with propolis extract (treatment group). In the treatment group, propolis was given 200 mg/kg per oral for 7 days then harvested brain tissues after sacrificed by cervical dislocation at day 8. We investigated Hsp70, Caspase 3, apoptosis-inducing factor (AIF), and TUNEL assay expression using immunohistochemistry staining. Statistical test using one-way ANOVA test and Tukey HSD as post hoc test. RESULTS: Mean of positive Hsp70 stained cells in group 1 was 6.82 ± 2.14, group 2 was 3.91 ± 2.26, and group 3 was 9.64 ± 3.53 with a significant difference of Hsp70 expression distribution within groups (p = 0.0001). Mean of positive caspase 3 stained cells in group 1 was 5.45 ± 2.30, group 2 was 13.82 ± 2.44, and group 3 was 7.03 ± 1.54 with a significant difference of caspase3 expression distribution within groups (p=0.0001). Mean of positive AIF stained cells in group 1 was 5.36 ± 2.11, group 2 was 12.82 ± 1.40, and group 3 was 8.09 ± 1.81 with a significant difference of AIF expression distribution within groups (p = 0.0001). Mean of positive TUNEL assay stained cells in group 1 was 4.82 ± 2.04, group 2 was 11.55 ± 1.51, and group 3 was 7.64 ± 1.96 with a significant difference of TUNEL test expression distribution within groups (p = 0.0001). CONCLUSION: Propolis may protect brain cell from apoptosis after injury by maintaining Hsp70 expression in addition to antioxidant and anti-inflammatory.
Collapse
Affiliation(s)
- Steven Tandean
- Department of Neurosurgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Iskandar Japardi
- Department of Neurosurgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Michael Lumintang Loe
- Department of Neurosurgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Wibi Riawan
- Department of Biochemistry, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Julius July
- Department of Neurosurgery, Faculty of Medicine, Universitas Pelita Harapan (UPH), Neuroscience Centre Siloam Hospital Lippo Village, Tanggerang, Indonesia
| |
Collapse
|
7
|
Borhani-Haghighi M, Kashani IR, Mohamadi Y, Pasbakhsh P. Embryonic intraventricular transplantation of neural stem cells augments inflammation-induced prenatal brain injury. J Chem Neuroanat 2018; 94:54-62. [PMID: 29959975 DOI: 10.1016/j.jchemneu.2018.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Prenatal brain injury results from undesirable circumstances during the embryonic development. Current endeavors for treating this complication are basically excluded to postnatal therapeutic approaches. Neural stem cell therapy has shown great promise for treating neurodevelopmental disorders. To our knowledge, this is the first study that investigates the therapeutic effect of in utero transplantation of neural stem cells (NSCs) in inflammation model of prenatal brain injury. METHODS To induce prenatal injury, time-mated C57BL6J mice were intraperitoneally injected with 50 μg/kg lipopolysaccharide (LPS(on the day 15 of gestation. In the treatment group, NSCs were transplanted into the lateral ventricle of embryos on day 17 of gestation. The expression of GFAP, Iba-1, Olig2, and NeuN were assessed by real time PCR and immunohistochemistry. Changes in IL-6, TNF-α and IL-10 cytokines level, and caspase 3 activity were evaluated in the cortex of pups. RESULTS Intrauterine transplanted NSCs homed to the brain cortex of offspring. Brain levels of pro-inflammatory cytokines showed a significant downward trend in the NSCs group. Furthermore, NSCs ameliorated inflammation-induced reactive microgliosis and astrogliosis as well as cellular degeneration. Apoptosis inhibition in the treated group was demonstrated by the decline in the caspase 3 activity and dark neurons. CONCLUSION This study suggests a promising prospect to initiate the treatment of prenatal brain injury before birth by intrauterine transplantation of NSCs.
Collapse
Affiliation(s)
- Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Mohamadi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Hosseini SM, Farahmandnia M, Razi Z, Delavari S, Shakibajahromi B, Sarvestani FS, Kazemi S, Semsar M. Combination cell therapy with mesenchymal stem cells and neural stem cells for brain stroke in rats. Int J Stem Cells 2015; 8:99-105. [PMID: 26019759 PMCID: PMC4445714 DOI: 10.15283/ijsc.2015.8.1.99] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 02/08/2015] [Indexed: 12/25/2022] Open
Abstract
Objectives Brain stroke is the second most important events that lead to disability and morbidity these days. Although, stroke is important, there is no treatment for curing this problem. Nowadays, cell therapy has opened a new window for treating central nervous system disease. In some previous studies the Mesenchymal stem cells and neural stem cells. In this study, we have designed an experiment to assess the combination cell therapy (Mesenchymal and Neural stem cells) effects on brain stroke. Method and Materials The Mesenchymal stem cells were isolated from adult rat bone marrow and the neural stem cells were isolated from ganglion eminence of rat embryo 14 days. The Mesenchymal stem cells were injected 1 day after middle cerebral artery occlusion (MCAO) and the neural stem cells transplanted 7 day after MCAO. After 28 days, the neurological outcomes and brain lesion volumes were evaluated. Also, the activity of Caspase 3 was assessed in different groups. Result The group which received combination cell therapy had better neurological examination and less brain lesion. Also the combination cell therapy group had the least Caspase 3 activity among the groups. Conclusions The combination cell therapy is more effective than Mesenchymal stem cell therapy and neural stem cell therapy separately in treating the brain stroke in rats.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran ; Stem Cell laboratory, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Farahmandnia
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Razi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Delavari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benafsheh Shakibajahromi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Sabet Sarvestani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepehr Kazemi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Semsar
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|