1
|
Li H, Zhang J, Ma K, Ji J, An C, Jiang H, Qu H, Tang R, Ren X, Du Y, Zhao Q. Advancements in the treatment of cerebral ischemia-reperfusion injury: Acupuncture combined with mesenchymal stem cells transplantation. Medicine (Baltimore) 2025; 104:e41075. [PMID: 39792753 PMCID: PMC11730110 DOI: 10.1097/md.0000000000041075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Cerebral ischemia-reperfusion injury (CIRI) constitutes a significant etiology of exacerbated cerebral tissue damage subsequent to intravenous thrombolysis and endovascular mechanical thrombectomy in patients diagnosed with acute ischemic stroke. The treatment of CIRI has been extensively investigated through a multitude of clinical studies. Acupuncture has been demonstrated to be effective in treating CIRI. Recent 5 years studies have identified potential mechanisms of acupuncture, including regulation of autophagy, promotion of angiogenesis, inhibition of inflammation and apoptosis, modulation of cell activation, neuroplasticity regulation, and promotion of nerve regeneration. The transplantation of mesenchymal stem cells (MSCs) can effectively suppress apoptosis, modulate immune responses, and enhance the proliferation and migration of endogenous neural stem cells (NSCs), thereby compensating for the NSCs deficiency following cerebral ischemia/reperfusion injury. The combination of acupuncture and MSCs transplantation demonstrates superiority over individual treatments, significantly enhancing the survival rate of MSCs. Moreover, it facilitates the secretion of various cytokines to promote their homing and differentiation into functional neurons, thereby providing a novel approach for clinical treatment of CIRI.
Collapse
Affiliation(s)
- Huan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiaxin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kewen Ma
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jie Ji
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chengfei An
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hailun Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Qu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruohan Tang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuesong Ren
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuzheng Du
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qi Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
2
|
Zhou L, Wang Z, Zhong Q, Song B, Wang Y, Guan T, Liu Q. Ultra-Low-Dose UV-C Photo-stimulation Promotes Neural Stem Cells Differentiation via Presenilin 1 Mediated Notch and β-Catenin Activation. Mol Neurobiol 2024; 61:9491-9506. [PMID: 38649660 DOI: 10.1007/s12035-024-04185-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Light-based photo-stimulation has demonstrated promising effects on stem cell behavior, particularly in optimizing neurogenesis. However, the precise parameters for achieving optimal results, including the wavelengths, light intensity, radiating energy, and underlying mechanisms, remain incompletely understood. In this study, we focused on utilizing ultraviolet-C (UV-C) at a specific wavelength of 254 nm, with an ultra-low dose at intensity of 330 μW/cm2 and a total energy of 594 mJ/cm2 per day over a period of seven days, to stimulate the proliferation and differentiation of mouse neural stem cells (NSCs). The results revealed that the application of ultra-low-dose UV-C yielded the most significant effect in promoting differentiation when compared to mixed ultraviolet (UV) and ultraviolet-A (UV-A) radiation at equivalent exposure levels. The mechanism exploration elucidated the role of Presenilin 1 in mediating the activation of β-catenin and Notch 1 by the UV-C treatment, both of which are key factors facilitating NSCs proliferation and differentiation. These findings introduce a novel approach employing ultra-low-dose UV-C for specifically enhancing NSC differentiation, as well as the underlying mechanism. It would contribute valuable insights into brain stimulation and neurogenesis modulation for various diseases, offering potential therapeutic avenues for further exploration.
Collapse
Affiliation(s)
- Lin Zhou
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Bio-functional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Zihan Wang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qiuling Zhong
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Bing Song
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yan Wang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Qian Liu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Bing J, Sun J, Zhao R, Sun L, Xi C, Liu J, Zhang X, Zeng S. The effects of Wnt, BMP, and Notch signaling pathways on cell proliferation and neural differentiation in a song control nucleus (HVC) of Lonchura striata. Dev Neurobiol 2023; 83:157-166. [PMID: 37433016 DOI: 10.1002/dneu.22920] [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: 11/23/2022] [Revised: 04/06/2023] [Accepted: 06/24/2023] [Indexed: 07/13/2023]
Abstract
There is obvious sexual dimorphism in the song control system of songbirds. In the higher vocal center (HVC), cell proliferation and neuronal differentiation contribute to the net addition of neurons. However, the mechanism underlying these changes is unclear. Given that Wnt, Bmp, and Notch pathways are involved in cell proliferation and neuronal differentiation, no reports are available to study the role of the three pathways in the song control system. To address the issue, we studied cell proliferation in the ventricle zone overlying the developing HVC and neural differentiation within the HVC of Bengalese finches (Lonchura striata) at posthatching day 15 when HVC progenitor cells are generated on a large scale and differentiate into neurons, after Wnt and Bmp pathways were activated by using a pharmacological agonist (LiCl) or Bmp4, respectively, and the Notch pathway was inhibited by an inhibitor (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester), DAPT). The results indicated that both cell proliferation and neural differentiation toward HVC neurons increased significantly after activation of the Wnt signaling pathway or inhibition of the Notch signaling pathway. Although cell proliferation was increased, neural differentiation was inhibited after treatment with Bmp4. There was obvious synergetic enhancement in the number of proliferating cells after the coregulation of two or three signaling pathways. In addition, synergetic enhancement was also found in the Wnt and Notch pathways in neural differentiation toward neurons within HVC. These results suggest that the three signaling pathways are involved in cell proliferation and neural differentiation of HVC.
Collapse
Affiliation(s)
- Jie Bing
- Beijing, Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Jing Sun
- Beijing, Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Rui Zhao
- Beijing, Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Lina Sun
- Beijing, Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Chao Xi
- Beijing, Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Jin Liu
- Beijing, Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| | - Xinwen Zhang
- Hainan, Institute of Science and Technology, Haikou, China
- College of Life Sciences, Hainan Normal University, Haikou, China
| | - Shaoju Zeng
- Beijing, Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing, China
| |
Collapse
|
4
|
Liu Q, Telezhkin V, Jiang W, Gu Y, Wang Y, Hong W, Tian W, Yarova P, Zhang G, Lee SMY, Zhang P, Zhao M, Allen ND, Hirsch E, Penninger J, Song B. Electric field stimulation boosts neuronal differentiation of neural stem cells for spinal cord injury treatment via PI3K/Akt/GSK-3β/β-catenin activation. Cell Biosci 2023; 13:4. [PMID: 36624495 PMCID: PMC9830810 DOI: 10.1186/s13578-023-00954-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Neural stem cells (NSCs) are considered as candidates for cell replacement therapy in many neurological disorders. However, the propensity for their differentiation to proceed more glial rather than neuronal phenotypes in pathological conditions limits positive outcomes of reparative transplantation. Exogenous physical stimulation to favor the neuronal differentiation of NSCs without extra chemical side effect could alleviate the problem, providing a safe and highly efficient cell therapy to accelerate neurological recovery following neuronal injuries. RESULTS With 7-day physiological electric field (EF) stimulation at 100 mV/mm, we recorded the boosted neuronal differentiation of NSCs, comparing to the non-EF treated cells with 2.3-fold higher MAP2 positive cell ratio, 1.6-fold longer neuronal process and 2.4-fold higher cells ratio with neuronal spontaneous action potential. While with the classical medium induction, the neuronal spontaneous potential may only achieve after 21-day induction. Deficiency of either PI3Kγ or β-catenin abolished the above improvement, demonstrating the requirement of the PI3K/Akt/GSK-3β/β-catenin cascade activation in the physiological EF stimulation boosted neuronal differentiation of NSCs. When transplanted into the spinal cord injury (SCI) modelled mice, these EF pre-stimulated NSCs were recorded to develop twofold higher proportion of neurons, comparing to the non-EF treated NSCs. Along with the boosted neuronal differentiation following transplantation, we also recorded the improved neurogenesis in the impacted spinal cord and the significantly benefitted hind limp motor function repair of the SCI mice. CONCLUSIONS In conclusion, we demonstrated physiological EF stimulation as an efficient method to boost the neuronal differentiation of NSCs via the PI3K/Akt/GSK-3β/β-catenin activation. Pre-treatment with the EF stimulation induction before NSCs transplantation would notably improve the therapeutic outcome for neurogenesis and neurofunction recovery of SCI.
Collapse
Affiliation(s)
- Qian Liu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China. .,School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.
| | - Vsevolod Telezhkin
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,School of Dental Sciences, Farmington Place, Newcastle University, Newcastle Upon Tyne, NE2 4BW, UK
| | - Wenkai Jiang
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu Gu
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK
| | - Yan Wang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Hong
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
| | - Weiming Tian
- Bio-X Centre, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150080, China
| | - Polina Yarova
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,School of Dental Sciences, Farmington Place, Newcastle University, Newcastle Upon Tyne, NE2 4BW, UK
| | - Gaofeng Zhang
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Peng Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Min Zhao
- Department of Ophthalmology and Vision Science, University of California at Davis, Sacramento, CA, 95616, USA
| | - Nicholas D Allen
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Josef Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC - Vienna BioCenter, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Bing Song
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China. .,School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.
| |
Collapse
|
5
|
Fan L, He M, Mo W, Yao Q, He M, Jiang J. miR-204-5p Inhibits the Proliferation and Differentiation of Fetal Neural Stem Cells by Targeting Wingless-Related MMTV Integration Site 2 to Regulate the Ephrin-A2/EphA7 Pathway. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is mainly resulted from perinatal asphyxia, which can be repaired by NSCs. miR-204-5p is claimed to impact the activity NSCs. Our research will probe the miR-204-5p function in oxygen-glucose deprivation (OGD)-treated NSCs. miR-204-5p level
was enhanced and WNT2 level was reduced in HIE rats. Rat NSCs were stimulated with OGD condition under the managing of mimic or inhibitor of miR-204-5p. The declined cell viability, enhanced apoptosis, downregulated Tuj1 and GFAP levels, and shortened total neurite length were observed in
OGD-treated NSCs, which were further aggravated by the mimic and rescued by the inhibitor of miR-204-5p. Furthermore, the inactivated WNT2 and Ephrin-A2/EphA7 signaling pathway in OGD-stimulated NSCs was further repressed by the mimic and rescued by the inhibitor of miR-204-5p. In addition,
WNT2 was confirmed as the targeting of miR-204-5p. Lastly, the function of miR-204-5p mimic on the proliferation, apoptosis, differentiation, WNT2 and Ephrin-A2/EphA7 signaling pathway in OGD-stimulated NSCs was abolished by HLY78, an activator of Wnt signaling. Collectively, miR-204-5p repressed
the growth and differentiation of fetal NSCs by targeting WNT2 to regulate the Ephrin-A2/EphA7 pathway.
Collapse
|
6
|
Wang Y, Liu C, Chen Y, Chen T, Han T, Xue L, Xu B. Systemically Silencing Long Non-coding RNAs Maclpil With Short Interfering RNA Nanoparticles Alleviates Experimental Ischemic Stroke by Promoting Macrophage Apoptosis and Anti-inflammatory Activation. Front Cardiovasc Med 2022; 9:876087. [PMID: 35600488 PMCID: PMC9120540 DOI: 10.3389/fcvm.2022.876087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/06/2022] [Indexed: 01/17/2023] Open
Abstract
Background Maclpil is a proinflammatory long non-coding RNA highly expressed on monocyte-derived macrophages in the ischemic brain. This study investigated the impact and the mechanisms of systemically delivering nanoparticle Maclpil short interfering RNA (siRNA) on experimental ischemic stroke in a mouse model. Methods Ischemic stroke (focal cerebral ischemia) was induced in male C57BL/6 mice through the middle cerebral artery occlusion. Three hours thereafter, mice were intravenously injected with Maclpil siRNA or scramble siRNA nanoparticles. Bone marrow cell-derived macrophages were transfected with Maclpil or scramble siRNA and subjected to oxygen glucose deprivation culture. The influence of silencing Maclpil on stroke outcomes, neuroinflammation, and macrophage fates was assessed via histology, flow cytometry, Western blotting, and quantitative PCR analysis. Results Three days following stroke induction, siRNA silencing Maclpil substantially reduced ischemic infarction size and improved neurological behaviors. Silencing Maclpil also markedly attenuated the accumulation of monocyte-derived macrophages, CD4+ T cells, and CD8+ T cells in the ischemic hemisphere without affecting microglia cellularity. Reciprocally, myeloid cells and both subsets of T cells were elevated in mouse peripheral blood following Maclpil siRNA treatment. Under oxygen glucose deprivation conditions that mimicked hypoxia and hypoglycemia in vitro, Maclpil siRNA silencing augmented macrophage apoptosis in conjunction with upregulation of proapoptotic Bax and caspase 3 expressions. siRNA knocking down Maclpil skewed macrophages from proinflammatory classical toward anti-inflammatory alternative activation as evidenced by increased arginase 1, Ym1, and Fizz1 and reduced inducible nitric oxide synthase, IL-1β, and TNF-α mRNA levels. Consistent with macrophage phenotype switching, silencing Maclpil by siRNA enhanced fatty acid oxidation as indicated by increased mRNA levels of 3 key metabolic enzymes (ACADM, ACADVL, and HADHA). Conclusion Systemically silencing Maclpil by siRNA nanoparticles attenuated experimental ischemic stroke by promoting macrophage apoptosis and anti-inflammatory alternative activation. Identifying and targeting Maclpil human homolog(s) may help develop a novel therapy for stroke clinical management.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Medical Research Center, Peking University Third Hospital, Beijing, China
- *Correspondence: Yan Wang,
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, China
| | - Yong Chen
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Tiffany Chen
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Tao Han
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Lixiang Xue
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Baohui Xu
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
7
|
Qi C, Zhang J, Wang Y, Lin M, Gao J, Lu H. Valproic acid enhances neurosphere formation in cultured rat embryonic cortical cells through TGFβ1 signaling. J Biomed Res 2022; 36:127-140. [PMID: 35387900 PMCID: PMC9002158 DOI: 10.7555/jbr.36.20210109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
This study aimed to investigate the effect and mechanism of valproic acid (VPA) on the neurosphere formation in rat embryonic cortical cells. We used free-floating neurosphere formation as a model system to evaluate the VPA on the proliferation of neural stem cells (NSCs). We found a time- and dose-dependent increase in neurosphere formation and NSC proliferation after VPA treatment. Further RNA-seq analysis demonstrated that the upregulated TGFβ1 signaling might attribute to the effect of VPA on the neurosphere formation and NSC proliferation. Consistently, the neurosphere formation and NSC proliferation were blocked by the treatment with SB431542, an inhibitor of TGFβ1 receptor. Moreover, in a coculture system, NSCs treated with VPA significantly reduced the oxygen-glucose deprivation-induced neuronal apoptosis. Taken together, our results showed that VPA could enhance neurosphere formation and NSC proliferation by activating TGFβ1, which might be a novel therapeutic strategy for neurological disorders.
Collapse
Affiliation(s)
- Cui Qi
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiaqi Zhang
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yuanyuan Wang
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Mingyan Lin
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jun Gao
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Rehabilitation Medicine, Jiangsu Shengze Hospital affiliated to Nanjing Medical University, Suzhou, Jiangsu 215228,China
- Jun Gao, Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu 211166, China. Tel: +86-25-86869347, E-mail:
| | - Haiying Lu
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
- Haiying Lu, Department of Neurology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, GulouDistrict, Nanjing, Jiangsu 210008, China. Tel: +86-25-83116827, E-mail:
| |
Collapse
|
8
|
Chen M, Wang F, Wang H. Silencing of lncRNA XLOC_035088 Protects Middle Cerebral Artery Occlusion-Induced Ischemic Stroke by Notch1 Signaling. J Neuropathol Exp Neurol 2021; 80:60-70. [PMID: 33236068 PMCID: PMC7749712 DOI: 10.1093/jnen/nlaa129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Ischemic stroke represents one of the leading causes of mortality worldwide and especially in developing countries. It is crucial for finding effective therapeutic targets that protect the brain against ischemic injury. Long noncoding RNAs (lncRNAs) have emerged as major regulators of neurological diseases, and clarifying their roles in cerebral ischemic injury may provide novel targets for the treatment of ischemic stroke. We aimed to investigate the role of lncRNA-XLOC_035088 in middle cerebral artery occlusion (MCAO)-induced rat brain injury and oxygen-glucose deprivation (OGD)-reperfusion treated hippocampal neurons. In our findings, we found that XLOC_035088 expression was significantly upregulated in OGD-reperfusion treated hippocampal neurons and in different brain regions of MCAO-treated rats. XLOC_035088 silencing protected against MCAO-induced ischemic brain injury in vivo and OGD-induced hippocampal neuronal apoptosis in vitro. Intrahippocampal silencing of XLOC_035088 significantly decreased brain XLOC_035088 expression, reduced brain infarct size, and improved neurological function through inhibiting NOTCH1 following derepression of presenilin 2 (PSEN2). Taken together, this study provides evidence that the lncRNA XLOC_035088/PSEN2/Notch1 axis is involved in the pathogenesis of ischemic brain injury, and presents a promising therapeutic route for ischemic stroke.
Collapse
Affiliation(s)
- Miao Chen
- From the Department of Neurology, Shidong Hospital, Affiliated to University of Shanghai for Science and Technology
| | - Feng Wang
- Department of Neurology, Shanghai Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine (FW), Shanghai, PR China
| | - Hairong Wang
- From the Department of Neurology, Shidong Hospital, Affiliated to University of Shanghai for Science and Technology
| |
Collapse
|
9
|
Liu Q, Jiao Y, Yang W, Gao B, Hsu DK, Nolta J, Russell M, Lyeth B, Zanto TP, Zhao M. Intracranial alternating current stimulation facilitates neurogenesis in a mouse model of Alzheimer's disease. Alzheimers Res Ther 2020; 12:89. [PMID: 32703308 PMCID: PMC7376967 DOI: 10.1186/s13195-020-00656-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/15/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Neurogenesis is significantly impaired in the brains of both human patients and experimental animal models of Alzheimer's disease (AD). Although deep brain stimulation promotes neurogenesis, it is an invasive technique that may damage neural circuitry along the path of the electrode. To circumvent this problem, we assessed whether intracranial electrical stimulation to the brain affects neurogenesis in a mouse model of Alzheimer's disease (5xFAD). METHODS AND RESULTS We used Ki67, Nestin, and doublecortin (DCX) as markers and determined that neurogenesis in both the subventricular zone (SVZ) and hippocampus were significantly reduced in the brains of 4-month-old 5xFAD mice. Guided by a finite element method (FEM) computer simulation to approximately estimate current and electric field in the mouse brain, electrodes were positioned on the skull that were likely to deliver stimulation to the SVZ and hippocampus. After a 4-week program of 40-Hz intracranial alternating current stimulation (iACS), neurogenesis indicated by expression of Ki67, Nestin, and DCX in both the SVZ and hippocampus were significantly increased compared to 5xFAD mice who received sham stimulation. The magnitude of neurogenesis was close to the wild-type (WT) age-matched unmanipulated controls. CONCLUSION Our results suggest that iACS is a promising, less invasive technique capable of effectively stimulating the SVZ and hippocampus regions in the mouse brain. Importantly, iACS can significantly boost neurogenesis in the brain and offers a potential treatment for AD.
Collapse
Affiliation(s)
- Qian Liu
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
- Center for Neuroscience, Department of Neurological Surgery, School of Medicine, University of California at Davis, Sacramento, CA, 95817, USA
| | - Yihang Jiao
- Department of Electrical and Computer Engineering, University of California at Davis, Davis, CA, 95616, USA
| | - Weijian Yang
- Department of Electrical and Computer Engineering, University of California at Davis, Davis, CA, 95616, USA
| | - Beiyao Gao
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
- Present location: Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, 200041, P. R. China
| | - Daniel K Hsu
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Jan Nolta
- Stem Cell Program and Gene Therapy Center, Institute for Regenerative Cures, Department of Internal Medicine, University of California at Davis, Sacramento, 95817, CA, USA
| | - Michael Russell
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Bruce Lyeth
- Center for Neuroscience, Department of Neurological Surgery, School of Medicine, University of California at Davis, Sacramento, CA, 95817, USA
| | - Theodore P Zanto
- Neuroscape, Department of Neurology, University of California San Francisco - Mission Bay, Sandler Neuroscience Center MC 0444, San Francisco, CA, 94158, USA.
| | - Min Zhao
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA.
- Center for Neuroscience, Department of Neurological Surgery, School of Medicine, University of California at Davis, Sacramento, CA, 95817, USA.
- Department of Ophthalmology and Vision Science, University of California at Davis, Sacramento, CA, 95616, USA.
| |
Collapse
|
10
|
Intraventricular Medium B Treatment Benefits an Ischemic Stroke Rodent Model via Enhancement of Neurogenesis and Anti-apoptosis. Sci Rep 2020; 10:6596. [PMID: 32313130 PMCID: PMC7171187 DOI: 10.1038/s41598-020-63598-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/31/2020] [Indexed: 12/26/2022] Open
Abstract
Enhancement of endogenous neurogenesis after ischemic stroke may improve functional recovery. We previously demonstrated that medium B, which is a combination with epidermal growth factor (EGF) and fibronectin, can promote neural stem/progenitor cell (NSPC) proliferation and migration. Here, we showed that medium B promoted proliferation and migration of cultured NSPCs onto various 3-dimentional structures. When rat cortical neurons with oxygen glucose deprivation (OGD) were co-cultured with NSPCs, medium B treatment increased neuronal viability and reduced cell apoptosis. In a rat model with transient middle cerebral artery occlusion (MCAO), post-insult intraventricular medium B treatment enhanced proliferation, migration, and neuronal differentiation of NSPCs and diminished cell apoptosis in the infarct brain. In cultured post-OGD neuronal cells and the infarct brain from MCAO rats, medium B treatment increased protein levels of Bcl-xL, Bcl-2, phospho-Akt, phospho-GSK-3β, and β-catenin and decreased the cleaved caspase-3 level, which may be associated with the effects of anti-apoptosis. Notably, intraventricular medium B treatment increased neuronal density, improved motor function and reduced infarct size in MCAO rats. In summary, medium B treatment results in less neuronal death and better functional outcome in both cellular and rodent models of ischemic stroke, probably via promotion of neurogenesis and reduction of apoptosis.
Collapse
|
11
|
Hu T, Sun R, Huang F, Liu X, Duan Z, Ye R, Li Y, Xiao L, Guo Z, Liu Q, Zhu W. CD99 mediates neutrophil transmigration through the bEnd.3 monolayer via the induction of oxygen-glucose deprivation. Biochem Biophys Res Commun 2020; 526:799-804. [PMID: 32268960 DOI: 10.1016/j.bbrc.2020.03.159] [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: 03/16/2020] [Accepted: 03/29/2020] [Indexed: 11/27/2022]
Abstract
AIM/BACKGROUND CD99 participate in neutrophil infiltration after inflammatory events; however, despite the important role of inflammation in ischemic stroke, the role of CD99 in ischemic stroke remains unclear. METHOD In the present study, we detected the protein expression of CD99, ICAM-1, and CD31 (PECAM-1) in oxygen-glucose deprivation (OGD)-induced bEnd.3 cells and neutrophils and explored the influence of HIF-1α and IL-1β on their expression. We also explored the role of CD99 in the OGD-induced transmigration of neutrophils. RESULTS Our results showed that OGD induction upregulated CD99 in bEnd.3 cells and that this effect could be abolished by the preadministration of IL-1β and was not mediated by HIF-1α. However, the activation of ICAM-1 by OGD remained activated with IL-1β treatment. No significant influence of IL-1β on OGD-induced CD31. Finally, we found a significant increase in infiltrated neutrophils after OGD induction compared with the control and OGD + anti-CD99 groups. CONCLUSION Our results indicated that CD99 mediates neutrophil infiltration and transmigration via OGD induction and thus constitutes a potential therapeutic target for anti-inflammatory treatment after ischemic stroke.
Collapse
Affiliation(s)
- Ting Hu
- Jinling Hospital Department of Neurology, Nanjing University, School of Medicine, Nanjing, 210002, PR China; Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China.
| | - Rui Sun
- Jinling Hospital Department of Neurology, Nanjing Medical University, Nanjing, 210002, PR China.
| | - Feihong Huang
- Nanjing Hospital Department of Neurology, South Medical University, Nanjing, 210002, PR China.
| | - Xiaoyun Liu
- Jinling Hospital Department of Neurology, Nanjing University, School of Medicine, Nanjing, 210002, PR China.
| | - Zhenhui Duan
- Nanjing Hospital Department of Neurology, South Medical University, Nanjing, 210002, PR China.
| | - Ruidong Ye
- Jinling Hospital Department of Neurology, Nanjing University, School of Medicine, Nanjing, 210002, PR China.
| | - Yunzi Li
- Jinling Hospital Department of Neurology, Nanjing University, School of Medicine, Nanjing, 210002, PR China.
| | - Lulu Xiao
- Jinling Hospital Department of Neurology, Nanjing University, School of Medicine, Nanjing, 210002, PR China.
| | - Zhiliang Guo
- Department of Neurology, Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| | - Qian Liu
- Jinling Hospital Department of Neurology, Nanjing University, School of Medicine, Nanjing, 210002, PR China.
| | - Wusheng Zhu
- Jinling Hospital Department of Neurology, Nanjing University, School of Medicine, Nanjing, 210002, PR China.
| |
Collapse
|
12
|
Li HM, Tong Y, Xia X, Huang J, Song PW, Zhang RJ, Shen CL. Retracted: Bone Mesenchymal Stem Cell-Conditioned Medium Regulates the Differentiation of Neural Stem Cells Via Notch Pathway Activation. Cell Reprogram 2018; 21:e339-e345. [PMID: 30589560 DOI: 10.1089/cell.2018.0042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The online-ahead-of print e-pub version of the article entitled, Bone Mesenchymal Stem Cell-Conditioned Medium Regulates the Differentiation of Neural Stem Cells Via Notch Pathway Activation, by Li H-M, Tong Y, Xia X, Huang J, Song P-W, Zhang R-J, Shen C-L, utilizing the DOI number 10.1089/cell.2018.0042 is being officially retracted from Cellular Reprogramming. The original version of the paper was submitted to the journal for peer review on July 29, 2018, with the revised version after peer review submitted on October 21, 2018. The paper was accepted for publication on November 20, 2018 and was subsequently published online ahead of print on December 27, 2018. After the e-publication of the article, the editor received an email from the corresponding author on January 14, 2019 requesting "to withdraw the above-mentioned manuscript for further consideration, due to a technical reason (we have done a further experiment and found this article need add more results)." Though it is unclear why the authors were not able to determine these faults with the paper within the six months the manuscript was in review, revision, and production, the editorial leadership of the Journal has determined that the paper requires a full retraction from the literature as Cellular Reprogramming is committed to upholding the strictest standards and best practices of scientific publishing.
Collapse
Affiliation(s)
- Hui-Min Li
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Yi Tong
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Xiang Xia
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Jian Huang
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Pei-Wen Song
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Ren-Jie Zhang
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Cai-Liang Shen
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Atorvastatin Rejuvenates Neural Stem Cells Injured by Oxygen–Glucose Deprivation and Induces Neuronal Differentiation Through Activating the PI3K/Akt and ERK Pathways. Mol Neurobiol 2018; 56:2964-2977. [DOI: 10.1007/s12035-018-1267-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 07/18/2018] [Indexed: 01/01/2023]
|
14
|
GRP78 Promotes Neural Stem Cell Antiapoptosis and Survival in Response to Oxygen-Glucose Deprivation (OGD)/Reoxygenation through PI3K/Akt, ERK1/2, and NF- κB/p65 Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3541807. [PMID: 29849883 PMCID: PMC5914129 DOI: 10.1155/2018/3541807] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/08/2018] [Accepted: 03/18/2018] [Indexed: 12/17/2022]
Abstract
When brain injury happens, endogenous neural stem cells (NSCs) located in the adult subventricular zone (SVZ) and subgranular zone (SGZ) are attacked by ischemia/reperfusion to undergo cellular apoptosis and death before being induced to migrate to the lesion point and differentiate into mature neural cells for damaged cell replacement. Although promoting antiapoptosis and NSC survival are critical to neuroregeneration, the mechanism has yet been elucidated clearly. Here in this study, we established an in vitro oxygen-glucose deprivation (OGD)/reoxygenation model on NSCs and detected glucose-regulated protein 78 (GRP78) involved in apoptosis, while in the absence of GRP78 by siRNA transfection, OGD/reoxygenation triggered PI3K/Akt, ERK1/2, and NF-κB/p65 activation, and induced NSC apoptosis was attenuated. Further investigation, respectively, with the inhibitor of PI3K/Akt or ERK1/2 demonstrated a blockage on GRP78 upregulation, while the inhibition of NF-κB rarely affected GRP78 induction by OGD/reoxygenation. The results indicated the bidirectional regulations of GRP78-PI3K/Akt and GRP78-ERK1/2 and the one-way signalling transduction through GRP78 to NF-κB/p65 on NSC survival from OGD/reoxygenation. In conclusion, we found that GRP78 mediated the signalling cross talk through PI3K/Akt, ERK1/2, and NF-κB/p65, which leads to antiapoptosis and NSC survival from ischemic stroke. Our finding gives a new evidence of GRP78 in NSCs as well as a new piece of signalling mechanism elucidation to NSC survival from ischemic stroke.
Collapse
|
15
|
Sfakis L, Kamaldinov T, Khmaladze A, Hosseini ZF, Nelson DA, Larsen M, Castracane J. Mesenchymal Cells Affect Salivary Epithelial Cell Morphology on PGS/PLGA Core/Shell Nanofibers. Int J Mol Sci 2018; 19:ijms19041031. [PMID: 29596382 PMCID: PMC5979364 DOI: 10.3390/ijms19041031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/21/2018] [Accepted: 03/28/2018] [Indexed: 12/15/2022] Open
Abstract
Engineering salivary glands is of interest due to the damaging effects of radiation therapy and the autoimmune disease Sjögren’s syndrome on salivary gland function. One of the current problems in tissue engineering is that in vitro studies often fail to predict in vivo regeneration due to failure of cells to interact with scaffolds and of the single cell types that are typically used for these studies. Although poly (lactic co glycolic acid) (PLGA) nanofiber scaffolds have been used for in vitro growth of epithelial cells, PLGA has low compliance and cells do not penetrate the scaffolds. Using a core-shell electrospinning technique, we incorporated poly (glycerol sebacate) (PGS) into PLGA scaffolds to increase the compliance and decrease hydrophobicity. PGS/PLGA scaffolds promoted epithelial cell penetration into the scaffold and apical localization of tight junction proteins, which is necessary for epithelial cell function. Additionally, co-culture of the salivary epithelial cells with NIH3T3 mesenchymal cells on PGS/PLGA scaffolds facilitated epithelial tissue reorganization and apical localization of tight junction proteins significantly more than in the absence of the mesenchyme. These data demonstrate the applicability of PGS/PLGA nanofibers for epithelial cell self-organization and facilitation of co-culture cell interactions that promote tissue self-organization in vitro.
Collapse
Affiliation(s)
- Lauren Sfakis
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA.
| | - Tim Kamaldinov
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA.
| | - Alexander Khmaladze
- Department of Physics, University at Albany, State University of New York, Albany, NY 12222, USA.
| | - Zeinab F Hosseini
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA.
| | - Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA.
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA.
| | - James Castracane
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA.
| |
Collapse
|
16
|
Inhibition of HSP90 Promotes Neural Stem Cell Survival from Oxidative Stress through Attenuating NF- κB/p65 Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3507290. [PMID: 27818721 PMCID: PMC5080492 DOI: 10.1155/2016/3507290] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 01/19/2023]
Abstract
Stem cell survival after transplantation determines the efficiency of stem cell treatment, which develops as a novel potential therapy for several central nervous system (CNS) diseases in recent decades. The engrafted stem cells face the damage of oxidative stress, inflammation, and immune response at the lesion point in host. Among the damaging pathologies, oxidative stress directs stem cells to apoptosis and even death through several signalling pathways and DNA damage. However, the in-detail mechanism of stem cell survival from oxidative stress has not been revealed clearly. Here, in this study, we used hydrogen peroxide (H2O2) to induce the oxidative damage on neural stem cells (NSCs). The damage was in consequence demonstrated involving the activation of heat shock protein 90 (HSP90) and NF-κB/p65 signalling pathways. Further application of the pharmacological inhibitors, respectively, targeting at each signalling indicated an upper-stream role of HSP90 upon NF-κB/p65 on NSCs survival. Preinhibition of HSP90 with the specific inhibitor displayed a significant protection on NSCs against oxidative stress. In conclusion, inhibition of HSP90 would attenuate NF-κB/p65 activation by oxidative induction and promote NSCs survival from oxidative damage. The HSP90/NF-κB mechanism provides a new evidence on rescuing NSCs from oxidative stress and also promotes the stem cell application on CNS pathologies.
Collapse
|
17
|
Cai Z, Zhao B, Deng Y, Shangguan S, Zhou F, Zhou W, Li X, Li Y, Chen G. Notch signaling in cerebrovascular diseases (Review). Mol Med Rep 2016; 14:2883-98. [PMID: 27574001 PMCID: PMC5042775 DOI: 10.3892/mmr.2016.5641] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/22/2016] [Indexed: 12/30/2022] Open
Abstract
The Notch signaling pathway is a crucial regulator of numerous fundamental cellular processes. Increasing evidence suggests that Notch signaling is involved in inflammation and oxidative stress, and thus in the progress of cerebrovascular diseases. In addition, Notch signaling in cerebrovascular diseases is associated with apoptosis, angiogenesis and the function of blood-brain barrier. Despite the contradictory results obtained to date as to whether Notch signaling is harmful or beneficial, the regulation of Notch signaling may provide a novel strategy for the treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Bin Zhao
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanqing Deng
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Shouqin Shangguan
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Faming Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wenqing Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiaoli Li
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanfeng Li
- Department of Neurology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Guanghui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
18
|
Li X, Bao X, Wang R. Neurogenesis-based epigenetic therapeutics for Alzheimer's disease (Review). Mol Med Rep 2016; 14:1043-53. [DOI: 10.3892/mmr.2016.5390] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 04/14/2016] [Indexed: 11/06/2022] Open
|
19
|
MAZ mediates the cross-talk between CT-1 and NOTCH1 signaling during gliogenesis. Sci Rep 2016; 6:21534. [PMID: 26867947 PMCID: PMC4751466 DOI: 10.1038/srep21534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 01/14/2016] [Indexed: 11/08/2022] Open
Abstract
Neurons and glia cells are differentiated from neural stem/progenitor cells (NSCs/NPCs) during brain development. Concomitant activation of JAK/STAT and NOTCH1 signaling is required for gliogenesis, a process to generate glia cells to ensure proper brain functions. NOTCH1 signaling is down-regulated during neurogenesis and up-regulated during gliogenesis. However, the underlying mechanism remains elusive. We report here that cardiotrophin-1 (CT-1) activates NOTCH1 signaling through the up-regulation of ADAM10, a rate-limiting factor of NOTCH1 signaling activation. We found that a transcriptional factor, Myc-associated zinc finger protein (MAZ), plays an important role in ADAM10 transcription in response to CT-1 in NPCs. MAZ knockdown inhibits CT-1 stimulated gliogenesis and it can be rescued by over-expressing human NICD. Our results provide a link between NOTCH1 activation and neuronal secreted CT-1, suggesting that CT-1 plays an important role in ensuring the coordinated activation of NOTCH1 signaling during gliogenesis.
Collapse
|
20
|
Li XY, Bao XJ, Wang RZ. Potential of neural stem cell-based therapies for Alzheimer's disease. J Neurosci Res 2015; 93:1313-24. [PMID: 25601591 DOI: 10.1002/jnr.23555] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/23/2014] [Accepted: 12/15/2014] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD), known to be a leading cause of dementia that causes heavy social and financial burdens worldwide, is characterized by progressive loss of neurons and synaptic connectivity after depositions of amyloid-β (Aβ) protein. Current therapies for AD patients can only alleviate symptoms but cannot deter the neural degeneration, thus providing no long-term recovery. Neural stem cells (NSCs), capable of self-renewal and of differentiation into functional neurons and glia, have been shown to repair damaged networks and reverse memory and learning deficits in animal studies, providing new hope for curing AD patients by cell transplantation. Under AD pathology, the microenvironment also undergoes great alterations that affect the propagation of NSCs and subsequent therapeutic efficiency, calling for measures to improve the hostile environment for cell transplantation. This article reviews the therapeutic potential of both endogenous and exogenous NSCs in the treatment of AD and the challenges to application of stem cells in AD treatment, particularly those from the microenvironmental alterations, in the hope of providing more information for future research in exploiting stem cell-based therapies for AD. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xue-Yuan Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Xin-Jie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Ren-Zhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|