1
|
Kathanadan Chackochan B, Johnson S, Thameemul Ansari HJ, Vengellur A, Sivan U, Koyyappurath S, P S BC. Transcriptomic analysis of CNTF-treated mouse subventricular zone-derived neurosphere culture reveals key transcription factor genes related to adult neurogenesis. Heliyon 2024; 10:e38496. [PMID: 39430537 PMCID: PMC11490819 DOI: 10.1016/j.heliyon.2024.e38496] [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: 03/04/2024] [Revised: 09/03/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
Neural Stem Progenitor Cells (NSPCs) maintenance and neuronal cell differentiation are the two key aspects of sustained neurogenesis in the adult mammalian brain. Transcription factors (TFs) are known to regulate these biological processes under the influence of various neurotrophic factors. Understanding the role of key TF genes in regulating adult neurogenesis is essential for determining the functional complexity and neuronal diversity seen in the adult mammalian brain. Although several molecular mechanisms leading to adult neurogenesis have been reported, details on its transcriptional regulation are still limited. Our initial results showed that Ciliary Neurotrophic Factor (CNTF) induced neuronal differentiation in SVZ-derived NSPC cultures. To investigate further the role of CNTF in inducing the expression of TF genes related to adult neurogenesis and the potential pathways involved, whole transcriptome RNA-sequencing (RNA-seq) analysis was done in CNTF-treated Sub-ventricular Zone derived neurosphere cultures from the mouse brain. The study revealed 483 differentially expressed genes (DEGs), among which 33 DEGs were identified as coding for transcription factors (TFs). Kyoto Encyclopedia of Gene and Genomes (KEGG) analysis revealed MAPK, PI3K-Akt, and FoxO as the significantly enriched signaling pathways. Gene co-expression network analysis identified five upregulated TF genes related to adult neurogenesis (Runx1, Hmga2, Fos, ID2, and Prrx1) in a single cluster, interacting with each other, and was also validated by quantitative PCR. Our data suggest several potential TFs that may act as critical regulators in the intrinsic transcriptional networks driving the adult neurogenesis process. Further investigation into these molecular regulators may yield a homogeneous population of neuronal progenitors for translational stem cell studies in the future.
Collapse
Affiliation(s)
- Bins Kathanadan Chackochan
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Sinoy Johnson
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Hilmi Jaufer Thameemul Ansari
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ajith Vengellur
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Unnikrishnan Sivan
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Kerala University of Fisheries and Ocean Studies, Cochin -682506, Kerala, India
| | - Sayuj Koyyappurath
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
| | - Baby Chakrapani P S
- Department of Biotechnology, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Neuroscience, Cochin University of Science and Technology, Cochin-682022, Kerala, India
- Centre for Excellence in Neurodegeneration and Brain Health, Kerala, India
| |
Collapse
|
2
|
Singh L, Bhatti R. Signaling Pathways Involved in the Neuroprotective Effect of Osthole: Evidence and Mechanisms. Mol Neurobiol 2024; 61:1100-1118. [PMID: 37682453 DOI: 10.1007/s12035-023-03580-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023]
Abstract
Neurodegenerative diseases constitute a major threat to human health and are usually accompanied by progressive structural and functional loss of neurons. Abnormalities in synaptic plasticity are involved in neurodegenerative disorders. Aberrant cell signaling cascades play a predominant role in the initiation, progress as well as in the severity of these ailments. Notch signaling is a pivotal role in the maintenance of neural stem cells and also participates in neurogenesis. PI3k/Akt cascade regulates different biological processes including cell proliferation, apoptosis, and metabolism. It regulates neurotoxicity and mediates the survival of neurons. Moreover, the activated BDNF/TrkB cascade is involved in promoting the transcription of genes responsible for cell survival and neurogenesis. Despite significant progress made in delineating the underlying pathological mechanisms involved and derangements in cellular metabolic promenades implicated in these diseases, satisfactory strategies for the clinical management of these ailments are yet to be achieved. Therefore, the molecules targeting these cell signaling cascades may emerge as useful leads in developing newer management strategies. Osthole is an important ingredient of traditional Chinese medicinal plants, often found in various plants of the Apiaceae family and has been observed to target these aforementioned mediators. Until now, no review has been aimed to discuss the possible molecular signaling cascades involved in osthole-mediated neuroprotection at one platform. The current review aimed to explore the interplay of various mediators and the modulation of the different molecular signaling cascades in osthole-mediated neuroprotection. This review could open new insights into research involving diseases of neuronal origin, especially the effect on neurodegeneration, neurogenesis, and synaptic plasticity. The articles gathered to compose the current review were extracted by using the PubMed, Scopus, Science Direct, and Web of Science databases. A methodical approach was used to integrate and discuss all published original reports describing the modulation of different mediators by osthole to confer neuroprotection at one platform to provide possible molecular pathways. Based on the inclusion and exclusion criteria, 32 articles were included in the systematic review. Moreover, literature evidence was also used to construct the biosynthetic pathway of osthole. The current review reveals that osthole promotes neurogenesis and neuronal functioning via stimulation of Notch, BDNF/Trk, and P13k/Akt signaling pathways. It upregulates the expression of various proteins, such as BDNF, TrkB, CREB, Nrf-2, P13k, and Akt. Activation of Wnt by osthole, in turn, regulates downstream GSK-1β to inhibit tau phosphorylation and β-catenin degradation to prevent neuronal apoptosis. The activation of Wnt and inhibition of oxidative stress, Aβ, and GSK-3β mediated β-catenin degradation by osthole might also be involved in mediating the protection against neurodegenerative diseases. Furthermore, it also inhibits neuroinflammation by suppressing MAPK/NF-κB-mediated transcription of genes involved in the generation of inflammatory cytokines and NLRP-3 inflammasomes. This review delineates the various underlying signaling pathways involved in mediating the neuroprotective effect of osthole. Modulation of Notch, BDNF/Trk, MAPK/NF-κB, and P13k/Akt signaling pathways by osthole confers protection against neurodegenerative diseases. The preclinical effects of osthole suggest that it could be a valuable molecule in inspiring the development of new drugs for the management of neurodegenerative diseases and demands clinical studies to explore its potential. An effort has been made to unify the varied mechanisms and target sites involved in the neuroprotective effect of osthole. The comprehensive description of the molecular pathways in the present work reflects its originality and thoroughness. The reviewed literature findings may be extrapolated to suggest the role of othole as a "biological response modifier" which contributes to neuroprotection through kinase modulatory, immunomodulatory, and anti-oxidative activity, which is documented even at lower doses. The current review attempts to emphasize the gaps in the existing literature which can be explored in the future.
Collapse
Affiliation(s)
- Lovedeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
- University Institute of Pharma Sciences, Chandigarh University, Mohali, 140413, Punjab, India.
| | - Rajbir Bhatti
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| |
Collapse
|
3
|
Takata S, Sakata-Haga H, Shimada H, Tsukada T, Sakai D, Shoji H, Tomosugi M, Nakamura Y, Ishigaki Y, Iizuka H, Hayashi Y, Hatta T. LIF-IGF Axis Contributes to the Proliferation of Neural Progenitor Cells in Developing Rat Cerebrum. Int J Mol Sci 2022; 23:13199. [PMID: 36361987 PMCID: PMC9659294 DOI: 10.3390/ijms232113199] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 09/10/2023] Open
Abstract
In rodent models, leukemia inhibitory factor (LIF) is involved in cerebral development via the placenta, and maternal immune activation is linked to psychiatric disorders in the child. However, whether LIF acts directly on neural progenitor cells (NPCs) remains unclear. This study performed DNA microarray analysis and quantitative RT-PCR on the fetal cerebrum after maternal intraperitoneal or fetal intracerebral ventricular injection of LIF at day 14.5 (E14.5) and determined that the expression of insulin-like growth factors (IGF)-1 and -2 was induced by LIF. Physiological IGF-1 and IGF-2 levels in fetal cerebrospinal fluid (CSF) increased from E15.5 to E17.5, following the physiological surge of LIF levels in CSF at E15.5. Immunostaining showed that IGF-1 was expressed in the cerebrum at E15.5 to E19.5 and IGF-2 at E15.5 to E17.5 and that IGF-1 receptor and insulin receptor were co-expressed in NPCs. Further, LIF treatment enhanced cultured NPC proliferation, which was reduced by picropodophyllin, an IGF-1 receptor inhibitor, even under LIF supplementation. Our findings suggest that IGF expression and release from the NPCs of the fetal cerebrum in fetal CSF is induced by LIF, thus supporting the involvement of the LIF-IGF axis in cerebral cortical development in an autocrine/paracrine manner.
Collapse
Affiliation(s)
- Sho Takata
- Department of Neurosurgery, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Hiromi Sakata-Haga
- Department of Anatomy, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Hiroki Shimada
- Department of Medical Science, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Tsuyoshi Tsukada
- Department of Neurosurgery, Saiseikai Toyama Hospital, Toyama 931-8533, Toyama, Japan
| | - Daisuke Sakai
- Department of Biology, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Hiroki Shoji
- Department of Biology, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Mitsuhiro Tomosugi
- Department of Anatomy, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Yuka Nakamura
- Department of Life Science, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Yasuhito Ishigaki
- Department of Life Science, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Hideaki Iizuka
- Department of Neurosurgery, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Yasuhiko Hayashi
- Department of Neurosurgery, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| |
Collapse
|
4
|
Baazaoui N, Iqbal K. Alzheimer's Disease: Challenges and a Therapeutic Opportunity to Treat It with a Neurotrophic Compound. Biomolecules 2022; 12:biom12101409. [PMID: 36291618 PMCID: PMC9599095 DOI: 10.3390/biom12101409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with an insidious onset and multifactorial nature. A deficit in neurogenesis and synaptic plasticity are considered the early pathological features associated with neurofibrillary tau and amyloid β pathologies and neuroinflammation. The imbalance of neurotrophic factors with an increase in FGF-2 level and a decrease in brain derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) in the hippocampus, frontal cortex and parietal cortex and disruption of the brain micro-environment are other characteristics of AD. Neurotrophic factors are crucial in neuronal differentiation, maturation, and survival. Several attempts to use neurotrophic factors to treat AD were made, but these trials were halted due to their blood-brain barrier (BBB) impermeability, short-half-life, and severe side effects. In the present review we mainly focus on the major etiopathology features of AD and the use of a small neurotrophic and neurogenic peptide mimetic compound; P021 that was discovered in our laboratory and was found to overcome the difficulties faced in the administration of the whole neurotrophic factor proteins. We describe pre-clinical studies on P021 and its potential as a therapeutic drug for AD and related neurodegenerative disorders. Our study is limited because it focuses only on P021 and the relevant literature; a more thorough investigation is required to review studies on various therapeutic approaches and potential drugs that are emerging in the AD field.
Collapse
Affiliation(s)
- Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
- Correspondence: ; Tel.: +1-718-494-5259; Fax: +1-718-494-1080
| |
Collapse
|
5
|
Li S, Yang Y, Li N, Li H, Xu J, Zhao W, Wang X, Ma L, Gao C, Ding Y, Ji X, Ren C. Limb Remote Ischemic Conditioning Promotes Neurogenesis after Cerebral Ischemia by Modulating miR-449b/Notch1 Pathway in Mice. Biomolecules 2022; 12:biom12081137. [PMID: 36009031 PMCID: PMC9405712 DOI: 10.3390/biom12081137] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Neurogenesis plays an important role in the prognosis of stroke patients and is known to be promoted by the activation of the Notch1 signaling pathway. Studies on the airway epithelium have shown that miR-449b represses the Notch pathway. The study aimed to investigate whether limb remote ischemic conditioning (LRIC) was able to promote neurogenesis in cerebral ischemic mice, and to investigate the role of the miR-449b/Notch1 pathway in LRIC-induced neuroprotection. Male C57BL/6 mice (22–25 g) were subjected to transient middle cerebral artery occlusion (MCAO), and LRIC was performed in the bilateral lower limbs immediately after MCA occlusion. Immunofluorescence staining was performed to assess neurogenesis. The cell line NE-4C was used to elucidate the proliferation of neuronal stem cells in 8% O2. After LRIC treatment on day 28, mice recovered neurological function. Neuronal precursor proliferation was enhanced in the SVZ, and neuronal precursor migration was enhanced in the basal ganglia on day 7. LRIC promoted the improvement of neurological function in mice on day 28, promoted neuronal precursor proliferation in the SVZ, and enhanced neuronal precursor migration in the basal ganglia on day 7. The neurological function score was negatively correlated with the number of BrdU-positive/DCX-positive cells in the SVZ and striatum. LRIC promoted activated Notch1 protein expression in the SVZ and substantially downregulated miR-449b levels in the SVZ and plasma. In vitro, miR-449b was found to target Notch1. Lentivirus-mediated miR-449b knockdown increased Notch1 levels in NE-4C cells and increased proliferation in the cells. The effects of miR-449b inhibition on neurogenesis were ablated by the application of Notch1 shRNA. Our study showed that LRIC promoted the proliferation and migration of neural stem cells after MCAO, and these effects were modulated by the miR-449b/Notch1 pathway.
Collapse
Affiliation(s)
- Sijie Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
- Emergency Department, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| | - Yong Yang
- School of Traditional Chinese Medicine, Beijing University of Chines Medicine, Beijing 100029, China
| | - Ning Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Haiyan Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
- School of Traditional Chinese Medicine, Beijing University of Chines Medicine, Beijing 100029, China
| | - Jiali Xu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Wenbo Zhao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Xiaojie Wang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518054, China
| | - Linqing Ma
- Department of Neurology, The People’s Hospital of Suzhou New District, Suzhou 215129, China
| | - Chen Gao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing 100053, China
- Correspondence: ; Tel.: +86-10-83198931; Fax: +86-10-63010085
| |
Collapse
|
6
|
Ye X, Li M, Bian W, Wu A, Zhang T, Li J, Zhou P, Cui H, Ding YQ, Liao M, Sun C. RBP-J deficiency promoted the proliferation and differentiation of CD133-positive cells in vitro and in vivo studies. Eur J Neurosci 2022; 56:3839-3860. [PMID: 35661443 DOI: 10.1111/ejn.15727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/02/2022] [Accepted: 05/29/2022] [Indexed: 11/27/2022]
Abstract
Although Notch signaling pathway could control the proliferation and differentiation of neural stem cells (NSCs), it is largely unknown about the effect of Notch signaling pathway on the neurogenesis of CD133-positive cells. By using the primary cultured ependymal cells and the transgenic mouse, we found that CD133 immunoreactivity was exclusively localized in the ependymal layer of ventricles, moreover, most CD133-positive cells were co-labeled with Nestin. In addition, recombination signal binding protein J (RBP-J), a key nuclear effector of Notch signaling pathway, was highly active in CD133-positive cells. CD133-positive cells can differentiate into the immature and mature neurons, in particular, the number of CD133-positive cells differentiating into the immature and mature neurons was significantly increased following the deficiency or interference of RBP-J in vivo or in vitro. By using real-time qPCR and western blot, we found that RBP-J and Hes1 were down-regulated while Notch1 was up-regulated in the expression levels of mRNAs and proteins following the deficiency or interference of RBP-J. These results demonstrated RBP-J deficiency promoted the proliferation and differentiation of CD133-positive cells. Therefore, we speculated that RBP-J could maintain CD133-positive cells in the characteristics of NSCs possibly by regulating Notch1/RBP-J/Hes1 pathway. It will provide a novel molecular insight into the function of RBP-J, as well as facilitate a future investigation of CD133-positive cells with respect to their potential application in neurodegenerative disorder.
Collapse
Affiliation(s)
- Xin Ye
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mengyi Li
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Bian
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Anting Wu
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ting Zhang
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Junwei Li
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Peng Zhou
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huairui Cui
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yu-Qiang Ding
- Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Min Liao
- Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Department of Histology and Embryology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chenyou Sun
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Institute of Neuroscience, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Dong C, Wang X, Sun L, Zhu L, Yang D, Gao S, Zhang W, Ling B, Liang A, Gao Z, Xu J. ATM modulates subventricular zone neural stem cell maintenance and senescence through Notch signaling pathway. Stem Cell Res 2021; 58:102618. [PMID: 34915311 DOI: 10.1016/j.scr.2021.102618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 09/22/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
Ataxia telangiectasia mutated (ATM) plays an essential role in DNA damage response and the maintenance of genomic stability. However, the role of ATM in regulating the function of adult neural stem cells (NSCs) remains unclear. Here we report that ATM deficiency led to accumulated DNA damage and decreased DNA damage repair capacity in neural progenitor cells. Moreover, we observed ATM ablation lead to the short-term increase of proliferation of neural progenitor cells, resulting in the depletion of the NSC pool over time, and this loss of NSC quiescence resulted in accelerated cell senescence. We further apply RNA sequencing to unravel that ATM knockout significantly affected Notch signaling pathway, furthermore, notch activation inhibit the abnormal increased proliferation of ATM-/- NSCs. Taken together, these findings indicate that ATM can serve as a key regulator for the normal function of adult NSCs by maintaining their stemness and preventing cellular senescence primarily through Notch signaling pathway.
Collapse
Affiliation(s)
- Chuanming Dong
- Department of Anatomy, Nantong University, Nantong 226001, China; East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xianli Wang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lixin Sun
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Liang Zhu
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Danjing Yang
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Shane Gao
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Wenjun Zhang
- Department of Hematology, Tongji Hospital of Tongji University School of Medicine, Shanghai 200065, China
| | - Bin Ling
- The Second People's Hospital of Yunnan Province, Kunming 650021, China.
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University School of Medicine, Shanghai 200065, China.
| | - Zhengliang Gao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
8
|
Kummer KK, Zeidler M, Kalpachidou T, Kress M. Role of IL-6 in the regulation of neuronal development, survival and function. Cytokine 2021; 144:155582. [PMID: 34058569 DOI: 10.1016/j.cyto.2021.155582] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
The pleiotropic cytokine interleukin-6 (IL-6) is emerging as a molecule with both beneficial and destructive potentials. It can exert opposing actions triggering either neuron survival after injury or causing neurodegeneration and cell death in neurodegenerative or neuropathic disorders. Importantly, neurons respond differently to IL-6 and this critically depends on their environment and whether they are located in the peripheral or the central nervous system. In addition to its hub regulator role in inflammation, IL-6 is recently emerging as an important regulator of neuron function in health and disease, offering exciting possibilities for more mechanistic insight into the pathogenesis of mental, neurodegenerative and pain disorders and for developing novel therapies for diseases with neuroimmune and neurogenic pathogenic components.
Collapse
Affiliation(s)
- Kai K Kummer
- Institute of Physiology, Medical University of Innsbruck, Austria
| | | | | | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Austria.
| |
Collapse
|
9
|
Chen J, Yuan XY, Zhang X. Intracerebral hemorrhage influences hippocampal neurogenesis and neurological function recovery via Notch1 signaling. Neuroreport 2021; 32:489-497. [PMID: 33657078 PMCID: PMC8016514 DOI: 10.1097/wnr.0000000000001614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/22/2020] [Indexed: 11/26/2022]
Abstract
Intracerebral hemorrhage (ICH) is associated with high rate of mortality and morbidity, but lacks effective therapies. Accumulating studies indicated that the hippocampal neurogenesis plays an essential role in the recovery of neurological function after ICH. The Notch1 signaling pathway shows important roles in neurogenesis. However, the effects of Notch1 on the recovery of neurological function after ICH remain unclear. Here, we used ICH mice model to investigate whether Notch1 signaling was involved in the hippocampal neurogenesis and the recovery of neurological function post-ICH. Our results showed that the rate of symmetric division pattern of hippocampal neural stem cells (NSCs) decreased significantly at 3 days after ICH. Meanwhile, the expression of Notch1 in the hippocampus also was reduced significantly. However, Notch1 activator treatment enhanced the expression of Notch1 and increased the number of Sox2+GFAP+ cells. Further, the rate of symmetric division pattern of NSCs also increased after Notch1 activator treatment in mice with ICH. Importantly, the number of DCX+ cells and BrdU+NeuN+ in hippocampus were increased on 28 days post-ICH as the Notch1 expression was upregulated. The motor function and spatial memory ability in post-ICH mice following Notch1 activator treatment also were improved. Taken together, our results suggested that Notch1 signaling could influence the recovery of long-term neurological function by regulating the proliferation and differentiation of the hippocampal NSCs in mice after ICH. Our study may provide ideas for the improvement of neurological function and spatial memory defects after ICH.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurology, Central Hospital of Baoji, Baoji
| | - Xing-Yun Yuan
- Department of Neurology, First Affiliated Hospital of Xi’an Jiao Tong University, Xian, Shanxi Province
| | - Xu Zhang
- Department of Cardiac Surgery, The General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Chen L, Wang Y, Chen Z. Adult Neurogenesis in Epileptogenesis: An Update for Preclinical Finding and Potential Clinical Translation. Curr Neuropharmacol 2021; 18:464-484. [PMID: 31744451 PMCID: PMC7457402 DOI: 10.2174/1570159x17666191118142314] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/31/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022] Open
Abstract
Epileptogenesis refers to the process in which a normal brain becomes epileptic, and is characterized by hypersynchronous spontaneous recurrent seizures involving a complex epileptogenic network. Current available pharmacological treatment of epilepsy is generally symptomatic in controlling seizures but is not disease-modifying in epileptogenesis. Cumulative evidence suggests that adult neurogenesis, specifically in the subgranular zone of the hippocampal dentate gyrus, is crucial in epileptogenesis. In this review, we describe the pathological changes that occur in adult neurogenesis in the epileptic brain and how adult neurogenesis is involved in epileptogenesis through different interventions. This is followed by a discussion of some of the molecular signaling pathways involved in regulating adult neurogenesis, which could be potential druggable targets for epileptogenesis. Finally, we provide perspectives on some possible research directions for future studies.
Collapse
Affiliation(s)
- Liying Chen
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Epilepsy Center, Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Epilepsy Center, Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Yong HJ, Ha N, Cho EB, Yun S, Kim H, Hwang JI, Seong JY. The unique expression profile of FAM19A1 in the mouse brain and its association with hyperactivity, long-term memory and fear acquisition. Sci Rep 2020; 10:3969. [PMID: 32123192 PMCID: PMC7052240 DOI: 10.1038/s41598-020-60266-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 02/10/2020] [Indexed: 12/30/2022] Open
Abstract
Neurodevelopment and mature brain function are spatiotemporally regulated by various cytokines and chemokines. The chemokine-like neuropeptide FAM19A1 is a member of family with sequence similarity 19 (FAM19), which is predominantly expressed in the brain. Its highly conserved amino acid sequence among vertebrates suggests that FAM19A1 may play important physiological roles in neurodevelopment and brain function. Here we used a LacZ reporter gene system to map the expression pattern of the FAM19A1 gene in the mouse brain. The FAM19A1 expression was observed in several brain regions starting during embryonic brain development. As the brain matured, the FAM19A1 expression was detected in the pyramidal cells of cortical layers 2/3 and 5 and in several limbic areas, including the hippocampus and the amygdala. FAM19A1-deficient mice were used to evaluate the physiological contribution of FAM19A1 to various brain functions. In behavior analysis, FAM19A1-deficient mice exhibited several abnormal behaviors, including hyperactive locomotor behavior, long-term memory deficits and fear acquisition failure. These findings provide insight into the potential contributions of FAM19A1 to neurodevelopment and mature brain function.
Collapse
Affiliation(s)
- Hyo Jeong Yong
- The GPCR laboratory, Graduate School of Biomedical Science, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Nui Ha
- Neuracle Science Co. Ltd., Seoul, 02841, Republic of Korea
| | - Eun Bee Cho
- Neuracle Science Co. Ltd., Seoul, 02841, Republic of Korea
| | - Seongsik Yun
- The GPCR laboratory, Graduate School of Biomedical Science, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Hyun Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jong-Ik Hwang
- The GPCR laboratory, Graduate School of Biomedical Science, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| | - Jae Young Seong
- The GPCR laboratory, Graduate School of Biomedical Science, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| |
Collapse
|
12
|
Anderson J, Patel M, Forenzo D, Ai X, Cai C, Wade Q, Risman R, Cai L. A novel mouse model for the study of endogenous neural stem and progenitor cells after traumatic brain injury. Exp Neurol 2020; 325:113119. [PMID: 31751572 PMCID: PMC10885014 DOI: 10.1016/j.expneurol.2019.113119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/23/2019] [Accepted: 11/16/2019] [Indexed: 11/29/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the US. Neural stem/progenitor cells (NSPCs) persist in the adult brain and represent a potential cell source for tissue regeneration and wound healing after injury. The Notch signaling pathway is critical for embryonic development and adult brain injury response. However, the specific role of Notch signaling in the injured brain is not well characterized. Our previous study has established a Notch1CR2-GFP reporter mouse line in which the Notch1CR2 enhancer directs GFP expression in NSPCs and their progeny. In this study, we performed closed head injury (CHI) in the Notch1CR2-GFP mice to study the response of injury-activated NSPCs. We show that CHI induces neuroinflammation, cell death, and the expression of typical TBI markers (e.g., ApoE, Il1b, and Tau), validating the animal model. In addition, CHI induces cell proliferation in GFP+ cells expressing NSPC markers, e.g., Notch1 and Nestin. A significant higher percentage of GFP+ astrocytes and GABAergic neurons was observed in the injured brain, with no significant change in oligodendrocyte lineage between the CHI and sham animal groups. Since injury is known to activate astrogliosis, our results suggest that injury-induced GFP+ NSPCs preferentially differentiate into GABAergic neurons. Our study establishes that Notch1CR2-GFP transgenic mouse is a useful tool for the study of NSPC behavior in vivo after TBI. Unveiling the potential of NSPCs response to TBI (e.g., proliferation and differentiation) will identify new therapeutic strategy for the treatment of brain trauma.
Collapse
Affiliation(s)
- Jeremy Anderson
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Misaal Patel
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Dylan Forenzo
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Xin Ai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Catherine Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Quinn Wade
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Rebecca Risman
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | - Li Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America.
| |
Collapse
|
13
|
Li J, Han Y, Li M, Nie C. Curcumin Promotes Proliferation of Adult Neural Stem Cells and the Birth of Neurons in Alzheimer's Disease Mice via Notch Signaling Pathway. Cell Reprogram 2019; 21:152-161. [PMID: 31145652 DOI: 10.1089/cell.2018.0027] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Jun Li
- Department of Neurology, Qinyang People's Hospital, Qinyang, China
| | - Yazhou Han
- Department of Neurology, Qinyang People's Hospital, Qinyang, China
| | - Mingduo Li
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Caixia Nie
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Daping Hospital & Institute of Surgery Research, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
14
|
Jia C, Keasey MP, Lovins C, Hagg T. Inhibition of astrocyte FAK-JNK signaling promotes subventricular zone neurogenesis through CNTF. Glia 2019; 66:2456-2469. [PMID: 30500112 DOI: 10.1002/glia.23498] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022]
Abstract
Astrocyte-derived ciliary neurotrophic factor (CNTF) promotes adult subventricular zone (SVZ) neurogenesis. We found that focal adhesion kinase (FAK) and JNK, but not ERK or P38, repress CNTF in vitro. Here, we defined the FAK-JNK pathway and its regulation of CNTF in mice, and the related leukemia inhibitory factor (LIF) and interleukin-6 (IL-6), which promote stem cell renewal at the expense of neurogenesis. Intrastriatal injection of FAK inhibitor, FAK14, in adult male C57BL/6 mice reduced pJNK and increased CNTF expression in the SVZ-containing periventricular region. Injection of a JNK inhibitor increased CNTF without affecting LIF and IL-6, and increased SVZ proliferation and neuroblast formation. The JNK inhibitor had no effect in CNTF-/- mice, suggesting that JNK inhibits SVZ neurogenesis by repressing CNTF. Inducible deletion of FAK in astrocytes increased SVZ CNTF and neurogenesis, but not LIF and IL-6. Intrastriatal injection of inhibitors suggested that P38 reduces LIF and IL-6 expression, whereas ERK induces CNTF and LIF. Intrastriatal FAK inhibition increased LIF, possibly through ERK, and IL-6 through another pathway that does not involve P38. Systemic injection of FAK14 also inhibited JNK while increasing CNTF, but did not affect P38 and ERK activation, or LIF and IL-6 expression. Importantly, systemic FAK14 increased SVZ neurogenesis in wild-type C57BL/6 and CNTF+/+ mice, but not in CNTF-/- littermates, indicating that it acts by upregulating CNTF. These data show a surprising differential regulation of related cytokines and identify the FAK-JNK-CNTF pathway as a specific target in astrocytes to promote neurogenesis and possibly neuroprotection in neurological disorders.
Collapse
Affiliation(s)
- Cuihong Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Matthew P Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Chiharu Lovins
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Theo Hagg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
15
|
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
|
16
|
Guidolin D, Fede C, Tortorella C. Nerve cells developmental processes and the dynamic role of cytokine signaling. Int J Dev Neurosci 2018; 77:3-17. [PMID: 30465872 DOI: 10.1016/j.ijdevneu.2018.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
The stunning diversity of neurons and glial cells makes possible the higher functions of the central nervous system (CNS), allowing the organism to sense, interpret and respond appropriately to the external environment. This cellular diversity derives from a single primary progenitor cell type initiating lineage leading to the formation of both differentiated neurons and glial cells. The processes governing the differentiation of the progenitor pool of cells into mature nerve cells will be here briefly reviewed. They involve morphological transformations, specialized modes of cell division, migration, and controlled cell death, and are regulated through cell-cell interactions and cues provided by the extracellular matrix, as well as by humoral factors from the cerebrospinal fluid and the blood system. In this respect, a quite large body of studies have been focused on cytokines, proteins representing the main signaling network that coordinates immune defense and the maintenance of homeostasis. At the same time, they are deeply involved in CNS development as regulatory factors. This dual role in the nervous system appears of particular relevance for CNS pathology, since cytokine dysregulation (occurring as a consequence of maternal infection, exposure to environmental factors or prenatal hypoxia) can profoundly impact on neurodevelopment and likely influence the response of the adult tissue during neuroinflammatory events.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, via Gabelli 65, I-35121, Padova, Italy
| | - Caterina Fede
- Department of Neuroscience, University of Padova, via Gabelli 65, I-35121, Padova, Italy
| | - Cinzia Tortorella
- Department of Neuroscience, University of Padova, via Gabelli 65, I-35121, Padova, Italy
| |
Collapse
|
17
|
Nasrolahi A, Mahmoudi J, Akbarzadeh A, Karimipour M, Sadigh-Eteghad S, Salehi R, Farhoudi M. Neurotrophic factors hold promise for the future of Parkinson's disease treatment: is there a light at the end of the tunnel? Rev Neurosci 2018; 29:475-489. [PMID: 29305570 DOI: 10.1515/revneuro-2017-0040] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 10/27/2017] [Indexed: 01/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and is characterized by a spectrum of clinicopathologic signs and a complex etiology. PD results from the degeneration of dopaminergic (DAergic) neurons in the substantia nigra. Current therapies for PD are only able to alleviate symptoms without stopping disease progression. In addition, the available therapeutic strategies do not have long-lasting effects. Furthermore, these therapies cause different ranges of adverse side effects. There is great interest in neurotrophic factors (NTFs) due to their ability to promote the survival of different neural cells. These factors are divided into four families: neurotrophins, neurokines, the glial cell line-derived NTF family of ligands, and the newly recognized cerebral DA NTF/mesencephalic astrocyte-derived NTF family. The protective and therapeutic effects of these factors on DAergic neurons make them suitable for the prevention of progressive cell loss in PD. Based on the above premise, we focus on the protective effects of NTFs, especially CDNF and MANF, on nigrostriatal DAergic neurons in PD.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Molecular Medicine Department, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran.,Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14756, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14756, Iran
| | - Abolfazl Akbarzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran
| | - Mohammad Karimipour
- Neuroscience Department, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran.,Department of Anatomy, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14756, Iran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14756, Iran.,Neuroscience Department, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran
| |
Collapse
|
18
|
Kitada M, Wakao S, Dezawa M. Intracellular signaling similarity reveals neural stem cell-like properties of ependymal cells in the adult rat spinal cord. Dev Growth Differ 2018; 60:326-340. [DOI: 10.1111/dgd.12546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 01/19/2023]
Affiliation(s)
- Masaaki Kitada
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology; Tohoku University Graduate School of Medicine; Sendai Japan
| |
Collapse
|
19
|
Yan Y, Kong L, Xia Y, Liang W, Wang L, Song J, Yao Y, Lin Y, Yang J. Osthole promotes endogenous neural stem cell proliferation and improved neurological function through Notch signaling pathway in mice acute mechanical brain injury. Brain Behav Immun 2018; 67:118-129. [PMID: 28823624 DOI: 10.1016/j.bbi.2017.08.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 01/19/2023] Open
Abstract
Mechanical brain injury (MBI) is a common neurotrosis disorder of the central nervous system (CNS), which has a higher mortality and disability. In the case of MBI, neurons death leads to loss of nerve function. To date, there was no satisfactory way to restore neural deficits caused by MBI. Endogenous neural stem cells (NSCs) can proliferate, differentiate and migrate to the lesions after brain injury, to replace and repair the damaged neural cells in the subventricular zone (SVZ), hippocampus and the regions of brain injury. In the present study, we first prepared a mouse model of cortical stab wound brain injury. Using the immunohistochemical and hematoxylin-eosin (H&E) staining method, we demonstrated that osthole (Ost), a natural coumarin derivative, was capable of promoting the proliferation of endogenous NSCs and improving neuronal restoration. Then, using the Morris water maze (MWM) test, we revealed that Ost significantly improved the learning and memory function in the MBI mice, increased the number of neurons in the regions of brain injury, hippocampus DG and CA3 regions. Additionally, we found that Ost up-regulated the expression of self-renewal genes Notch 1 and Hes 1. However, when Notch activity was blocked by the γ-secretase inhibitor DAPT, the expression of Notch 1 and Hes 1 mRNA was down-regulated, augmentation of NICD and Hes 1 protein was ameliorated, the proliferation-inducing effect of Ost was abolished. These results suggested that the effects of Ost were at least in part mediated by activation of Notch signaling pathway. Our findings support that Ost is a potential drug for treating MBI due to its neuronal restoration.
Collapse
Affiliation(s)
- Yuhui Yan
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, PR China
| | - Liang Kong
- China First Mandarin Group Northeast International Hospital, Shenyang 110623, PR China
| | - Yang Xia
- Department of Engineering, University of Oxford, Oxford OX1 3LZ, UK
| | - Wenbo Liang
- School of Medicine, Dalian University, Dalian 116622, PR China
| | - Litong Wang
- Department of Neurological Rehabilitation, The Second Affiliated Hospital of Dalian Medical University, Dalian 116600, Liaoning, PR China
| | - Jie Song
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, PR China
| | - Yingjia Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, PR China
| | - Ying Lin
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, PR China
| | - Jingxian Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, PR China.
| |
Collapse
|
20
|
Ludwig PE, Thankam FG, Patil AA, Chamczuk AJ, Agrawal DK. Brain injury and neural stem cells. Neural Regen Res 2018; 13:7-18. [PMID: 29451199 PMCID: PMC5840995 DOI: 10.4103/1673-5374.224361] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2017] [Indexed: 12/26/2022] Open
Abstract
Many therapies with potential for treatment of brain injury have been investigated. Few types of cells have spurred as much interest and excitement as stem cells over the past few decades. The multipotentiality and self-renewing characteristics of stem cells confer upon them the capability to regenerate lost tissue in ischemic or degenerative conditions as well as trauma. While stem cells have not yet proven to be clinically effective in many such conditions as was once hoped, they have demonstrated some effects that could be manipulated for clinical benefit. The various types of stem cells have similar characteristics, and largely differ in terms of origin; those that have differentiated to some extent may exhibit limited capability in differentiation potential. Stem cells can aid in decreasing lesion size and improving function following brain injury.
Collapse
Affiliation(s)
- Parker E. Ludwig
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Finosh G. Thankam
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Arun A. Patil
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
- Department of Neurosurgery, Creighton University School of Medicine, Omaha, NE, USA
| | - Andrea J. Chamczuk
- Department of Neurosurgery, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
21
|
Neuroinflammation and physical exercise as modulators of adult hippocampal neural precursor cell behavior. Rev Neurosci 2017; 29:1-20. [DOI: 10.1515/revneuro-2017-0024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022]
Abstract
Abstract
The dentate gyrus of the hippocampus is a plastic structure where adult neurogenesis constitutively occurs. Cell components of the neurogenic niche are source of paracrine as well as membrane-bound factors such as Notch, Bone Morphogenetic Proteins, Wnts, Sonic Hedgehog, cytokines, and growth factors that regulate adult hippocampal neurogenesis and cell fate decision. The integration and coordinated action of multiple extrinsic and intrinsic cues drive a continuous decision process: if adult neural stem cells remain quiescent or proliferate, if they take a neuronal or a glial lineage, and if new cells proliferate, undergo apoptotic death, or survive. The proper balance in the molecular milieu of this neurogenic niche leads to the production of neurons in a higher rate as that of astrocytes. But this rate changes in face of microenvironment modifications as those driven by physical exercise or with neuroinflammation. In this work, we first review the cellular and molecular components of the subgranular zone, focusing on the molecules, active signaling pathways and genetic programs that maintain quiescence, induce proliferation, or promote differentiation. We then summarize the evidence regarding the role of neuroinflammation and physical exercise in the modulation of adult hippocampal neurogenesis with emphasis on the activation of progression from adult neural stem cells to lineage-committed progenitors to their progeny mainly in murine models.
Collapse
|
22
|
Kuroda A, Fuchigami T, Fuke S, Koyama N, Ikenaka K, Hitoshi S. Minocycline Directly Enhances the Self-Renewal of Adult Neural Precursor Cells. Neurochem Res 2017; 43:219-226. [PMID: 29081002 DOI: 10.1007/s11064-017-2422-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/28/2017] [Accepted: 10/19/2017] [Indexed: 02/07/2023]
Abstract
Minocycline not only has antibacterial action but also produces a variety of pharmacological effects. It has drawn considerable attention as a therapeutic agent for symptoms caused by inflammation in many neurological disorders, leading to several clinical trials. Although some of these effects are mediated through its function of suppressing microglial activation, it is not clear whether minocycline acts on other cell types in the adult brain. In this study, we utilized a colony-forming neurosphere assay, in which neural stem cells (NSCs) clonally proliferate to form floating colonies, called neurospheres. We found that minocycline (at therapeutically relevant concentrations in cerebrospinal fluid) enhances the self-renewal capability of NSCs derived from the subependymal zone of adult mouse brain and facilitates their differentiation into oligodendrocytes. Importantly, these effects were independent of a suppression of microglial activation and were specifically observed with minocycline (among tetracycline derivatives). In addition, the size of the NSC population in the adult brain was increased when minocycline was infused into the lateral ventricle by an osmotic minipump in vivo. While precise molecular mechanisms of how minocycline alters the behavior of adult NSCs remain unknown, our data provide a basis for the clinical use of minocycline to treat neurodegenerative and demyelinating diseases.
Collapse
Affiliation(s)
- Anri Kuroda
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Takahiro Fuchigami
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Satoshi Fuke
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Natsu Koyama
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan.,Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies, Okazaki, 444-8787, Japan
| | - Seiji Hitoshi
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, 520-2192, Japan. .,Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan. .,Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies, Okazaki, 444-8787, Japan.
| |
Collapse
|
23
|
Davis SM, Pennypacker KR. The role of the leukemia inhibitory factor receptor in neuroprotective signaling. Pharmacol Ther 2017; 183:50-57. [PMID: 28827150 DOI: 10.1016/j.pharmthera.2017.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Several neurotropic cytokines relay their signaling through the leukemia inhibitory factor receptor. This 190kDa subunit couples with the 130kDa gp130 subunit to transduce intracellular signaling in neurons and oligodendrocytes that leads to expression of genes associated with neurosurvival. Moreover, activation of this receptor alters the phenotype of immune cells to an anti-inflammatory one. Although cytokines that activate the leukemia inhibitory factor receptor have been studied in the context of neurodegenerative disease, therapeutic targeting of the specific receptor subunit has been understudied in by comparison. This review examines the role of this receptor in the CNS and immune system, and its application in the treatment in stroke and other brain pathologies.
Collapse
Affiliation(s)
- Stephanie M Davis
- Center for Advanced Translational Stroke Science, Departments of Neurology and Neuroscience, University of Kentucky, Lexington, KY 40536, United States
| | - Keith R Pennypacker
- Center for Advanced Translational Stroke Science, Departments of Neurology and Neuroscience, University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|
24
|
Felling RJ, Covey MV, Wolujewicz P, Batish M, Levison SW. Astrocyte-produced leukemia inhibitory factor expands the neural stem/progenitor pool following perinatal hypoxia-ischemia. J Neurosci Res 2016; 94:1531-1545. [PMID: 27661001 PMCID: PMC5082180 DOI: 10.1002/jnr.23929] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 12/14/2022]
Abstract
Brain injuries, such as cerebral hypoxia-ischemia (H-I), induce a regenerative response from the neural stem/progenitors (NSPs) of the subventricular zone (SVZ); however, the mechanisms that regulate this expansion have not yet been fully elucidated. The Notch- Delta-Serrate-Lag2 (DSL) signaling pathway is considered essential for the maintenance of neural stem cells, but it is not known if it is necessary for the expansion of the NSPs subsequent to perinatal H-I injury. Therefore, the aim of this study was to investigate whether this pathway contributes to NSP expansion in the SVZ after H-I and, if so, to establish whether this pathway is directly induced by H-I or regulated by paracrine factors. Here we report that Notch1 receptor induction and one of its ligands, Delta-like 1, precedes NSP expansion after perinatal H-I in P6 rat pups and that this increase occurs specifically in the most medial cell layers of the SVZ where the stem cells reside. Pharmacologically inhibiting Notch signaling in vivo diminished NSP expansion. With an in vitro model of H-I, Notch1 was not induced directly by hypoxia, but was stimulated by soluble factors, specifically leukemia inhibitory factor, produced by astrocytes within the SVZ. These data confirm the importance both of the Notch-DSL signaling pathway in the expansion of NSPs after H-I and in the role of the support cells in their niche. They further support the body of evidence that indicates that leukemia inhibitory factor is a key injury-induced cytokine that is stimulating the regenerative response of the NSPs. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ryan J Felling
- Departments of Neurology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pharmacology, Physiology and Neuroscience, RBHS-New Jersey Medical School, Newark, New Jersey
| | - Matthew V Covey
- Department of Pharmacology, Physiology and Neuroscience, RBHS-New Jersey Medical School, Newark, New Jersey
| | - Paul Wolujewicz
- Department of Microbiology, Biochemistry and Molecular Genetics, RBHS-New Jersey Medical School, Newark, New Jersey
| | - Mona Batish
- Department of Microbiology, Biochemistry and Molecular Genetics, RBHS-New Jersey Medical School, Newark, New Jersey
| | - Steven W Levison
- Department of Pharmacology, Physiology and Neuroscience, RBHS-New Jersey Medical School, Newark, New Jersey.
| |
Collapse
|
25
|
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
|
26
|
Fonken LK, Gaudet AD, Gaier KR, Nelson RJ, Popovich PG. MicroRNA-155 deletion reduces anxiety- and depressive-like behaviors in mice. Psychoneuroendocrinology 2016; 63:362-9. [PMID: 26555429 DOI: 10.1016/j.psyneuen.2015.10.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/28/2015] [Accepted: 10/28/2015] [Indexed: 12/26/2022]
Abstract
Depressive disorders have complex and multi-faceted underlying mechanisms, rendering these disorders difficult to treat consistently and effectively. One under-explored therapeutic strategy for alleviating mood disorders is the targeting of microRNAs (miRs). miRs are small non-coding RNAs that cause sequestration/degradation of specific mRNAs, thereby preventing protein translation and downstream functions. miR-155 has validated and predicted neurotrophic factor and inflammatory mRNA targets, which led to our hypothesis that miR-155 deletion would modulate affective behaviors. To evaluate anxiety-like behavior, wildtype (wt) and miR-155 knockout (ko) mice (littermates; both male and female) were assessed in the open field and on an elevated plus maze. In both tests, miR-155 ko mice spent more time in open areas, suggesting they had reduced anxiety-like behavior. Depressive-like behaviors were assessed using the forced swim test. Compared to wt mice, miR-155 ko mice exhibited reduced float duration and increased latency to float. Further, although all mice exhibited a strong preference for a sucrose solution over water, this preference was enhanced in miR-155 ko mice. miR-155 ko mice had no deficiencies in learning and memory (Barnes maze) or social preference/novelty suggesting that changes in mood were specific. Finally, compared to wt hippocampi, miR-155 ko hippocampi had a reduced inflammatory signature (e.g., decreased IL-6, TNF-a) and female miR-155 ko mice increased ciliary neurotrophic factor expression. Together, these data highlight the importance of studying microRNAs in the context of anxiety and depression and identify miR-155 as a novel potential therapeutic target for improving mood disorders.
Collapse
Affiliation(s)
- Laura K Fonken
- Department of Neuroscience, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Columbus, OH 43210, USA.
| | - Andrew D Gaudet
- Department of Neuroscience, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | | | - Randy J Nelson
- Department of Neuroscience, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Columbus, OH 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
27
|
The effects of acute and chronic administration of phosphatidylserine on cell proliferation and survival in the dentate gyrus of adult and middle-aged rats. Brain Res 2015; 1609:72-81. [DOI: 10.1016/j.brainres.2015.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 03/04/2015] [Accepted: 03/09/2015] [Indexed: 11/19/2022]
|
28
|
Influences of prenatal and postnatal stress on adult hippocampal neurogenesis: the double neurogenic niche hypothesis. Behav Brain Res 2014; 281:309-17. [PMID: 25546722 DOI: 10.1016/j.bbr.2014.12.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 01/07/2023]
Abstract
Adult hippocampal neurogenesis (AHN) is involved in learning, memory, and stress, and plays a significant role in neurodegenerative and psychiatric disorders. As an age-dependent process, AHN is largely influenced by changes that occur during the pre- and postnatal stages of brain development, and constitutes an important field of research. This review examines the current knowledge regarding the regulators of AHN and the influence of prenatal and postnatal stress on later AHN. In addition, a hypothesis is presented suggesting that each kind of stress influences a specific neurogenic pool, developmental or postnatal, that later becomes a precursor with important repercussions for AHN. This hypothesis is referred to as "the double neurogenic niche hypothesis." Discovering what receptors, transcription factors, or genes are specifically activated by different stressors is proposed as an essential line of future research in the field. Such knowledge shall constitute an important starting point toward the goal of modifying AHN in neurodegenerative or psychiatric diseases.
Collapse
|
29
|
Abstract
Pathological ventricle remodelling, which follows a cardiac insult, causes heart failure. Despite the existence of multiple pharmaceutical approaches, heart failure is one of the leading causes of death worldwide and there is an urgent need to explore new therapeutic avenues. The Notch pathway is an evolutionary conserved fundamental pathway that regulates cell fate during development as well as throughout postnatal life in self-renewing tissues. In the myocardium, Notch signalling is involved in the modulation of cardiomyocytes survival, cardiac stem cells differentiation, and angiogenesis which are factors known to determine the extent of pathological cardiac remodelling. Modulation of the Notch pathway could become a tool to limit ventricle remodelling and the associated inexorable deterioration of cardiac performance.
Collapse
Affiliation(s)
- Roberto Ferrari
- Department of Cardiology and LTTA Centre, University Hospital of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- GVM Care and Research, E.S: Health Science Foundation, Maria Cecilia Hospital, Cotignola, Italy
| |
Collapse
|
30
|
Bolognin S, Buffelli M, Puoliväli J, Iqbal K. Rescue of cognitive-aging by administration of a neurogenic and/or neurotrophic compound. Neurobiol Aging 2014; 35:2134-46. [PMID: 24702821 DOI: 10.1016/j.neurobiolaging.2014.02.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/18/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
Aging is characterized by a progressive decline of cognitive performance, which has been partially attributed to structural and functional alterations of hippocampus. Importantly, aging is the major risk factor for the development of neurodegenerative diseases, especially Alzheimer's disease. An important therapeutic approach to counteract the age-associated memory dysfunctions is to maintain an appropriate microenvironment for successful neurogenesis and synaptic plasticity. In this study, we show that chronic oral administration of peptide 021 (P021), a small peptidergic neurotrophic compound derived from the ciliary neurotrophic factor, significantly reduced the age-dependent decline in learning and memory in 22 to 24-month-old Fisher rats. Treatment with P021 inhibited the deficit in neurogenesis in the aged rats and increased the expression of brain derived neurotrophic factor. Furthermore, P021 restored synaptic deficits both in the cortex and the hippocampus. In vivo magnetic resonance spectroscopy revealed age-dependent alterations in hippocampal content of several metabolites. Remarkably, P021 was effective in significantly reducing myoinositol (INS) concentration, which was increased in aged compared with young rats. These findings suggest that stimulating endogenous neuroprotective mechanisms is a potential therapeutic approach to cognitive aging, Alzheimer's disease, and associated neurodegenerative disorders and P021 is a promising compound for this purpose.
Collapse
Affiliation(s)
- Silvia Bolognin
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Mario Buffelli
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Jukka Puoliväli
- Department of Behavioral Studies, Charles River Finland, Kuopio, Finland
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA.
| |
Collapse
|
31
|
Kotasová H, Procházková J, Pacherník J. Interaction of Notch and gp130 signaling in the maintenance of neural stem and progenitor cells. Cell Mol Neurobiol 2014; 34:1-15. [PMID: 24132391 PMCID: PMC11488917 DOI: 10.1007/s10571-013-9996-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/30/2013] [Indexed: 01/10/2023]
Abstract
Notch and gp130 signaling are involved in the regulation of multiple cellular processes across various tissues during animal ontogenesis. In the developing nervous system, both signaling pathways intervene at many stages to determine cell fate-from the first neural lineage commitment and generation of neuronal precursors, to the terminal specification of cells as neurons and glia. In most cases, the effects of Notch and gp130 signaling in these processes are similar. The aim of the current review was to summarize the knowledge regarding the roles of Notch and gp130 signaling in the maintenance of neural stem and progenitor cells during animal ontogenesis, from early embryo to adult. Recent data show a direct crosstalk between these signaling pathways that seems to be specific for a particular type of neural progenitors.
Collapse
Affiliation(s)
- Hana Kotasová
- Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czech Republic
| | - Jiřina Procházková
- Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czech Republic
| | - Jiří Pacherník
- Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czech Republic
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
32
|
Sun F, Mao X, Xie L, Ding M, Shao B, Jin K. Notch1 signaling modulates neuronal progenitor activity in the subventricular zone in response to aging and focal ischemia. Aging Cell 2013; 12:978-87. [PMID: 23834718 DOI: 10.1111/acel.12134] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2013] [Indexed: 12/21/2022] Open
Abstract
Neurogenesis diminishes with aging and ischemia-induced neurogenesis also occurs, but reduced in aged brain. Currently, the cellular and molecular pathways mediating these effects remain largely unknown. Our previous study has shown that Notch1 signaling regulates neurogenesis in subventricular zone (SVZ) of young adult brain after focal ischemia, but whether a similar effect occurs in aged normal and ischemic animals is unknown. Here, we used normal and ischemic aged rat brains to investigate whether Notch1 signaling was involved in the reduction of neurogenesis in response to aging and modulates neurogenesis in aged brains after focal ischemia. By Western blot, we found that Notch1 and Jagged1 expression in the SVZ of aged brain was significantly reduced compared with young adult brain. Consistently, the activated form of Notch1 (Notch intracellular domain; NICD) expression was also declined. Immunohistochemistry confirmed that expression and activation of Notch1 signaling in the SVZ of aged brain were reduced. Double or triple immunostaining showed that that Notch1 was mainly expressed in doublecortin (DCX)-positive cells, whereas Jagged1 was predominantly expressed in astroglial cells in the SVZ of normal aged rat brain. In addition, disruption or activation of Notch1 signaling altered the number of proliferating cells labeled by bromodeoxyuridine (BrdU) and DCX in the SVZ of aged brain. Moreover, ischemia-induced cell proliferation in the SVZ of aged brain was enhanced by activating the Notch1 pathway and was suppressed by inhibiting the Notch1 signaling. Reduced infarct volume and improved motor deficits were also observed in Notch1 activator-treated aged ischemic rats. Our data suggest that Notch1 signaling modulates the SVZ neurogenesis in aged brain in normal and ischemic conditions.
Collapse
Affiliation(s)
- Fen Sun
- Department of Neurology; Second Affiliated Hospital; School of Medicine; Zhejiang University; No. 88 Jiefang Road Hangzhou Zhejiang 310009 China
- Department of Pharmacology & Neuroscience; Institute for Aging and Alzheimer's Disease Research; University of North Texas Health Science Center; 3500 Camp Bowie Boulevard Fort Worth TX 76107 USA
| | - XiaoOu Mao
- Buck Institute for Research on Aging; 8001 Redwood Blvd. Novato CA 94945 USA
| | - Lin Xie
- Buck Institute for Research on Aging; 8001 Redwood Blvd. Novato CA 94945 USA
| | - Meiping Ding
- Department of Neurology; Second Affiliated Hospital; School of Medicine; Zhejiang University; No. 88 Jiefang Road Hangzhou Zhejiang 310009 China
| | - Bei Shao
- Department of Neurology; First Affiliated Hospital; Wenzhou Medical University; 2 Fuxue Road Wenzhou Zhejiang 325000 China
| | - Kunlin Jin
- Department of Pharmacology & Neuroscience; Institute for Aging and Alzheimer's Disease Research; University of North Texas Health Science Center; 3500 Camp Bowie Boulevard Fort Worth TX 76107 USA
- Buck Institute for Research on Aging; 8001 Redwood Blvd. Novato CA 94945 USA
| |
Collapse
|
33
|
de Oliveira-Carlos V, Ganz J, Hans S, Kaslin J, Brand M. Notch receptor expression in neurogenic regions of the adult zebrafish brain. PLoS One 2013; 8:e73384. [PMID: 24039926 PMCID: PMC3767821 DOI: 10.1371/journal.pone.0073384] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 07/22/2013] [Indexed: 12/21/2022] Open
Abstract
The adult zebrash brain has a remarkable constitutive neurogenic capacity. The regulation and maintenance of its adult neurogenic niches are poorly understood. In mammals, Notch signaling is involved in stem cell maintenance both in embryonic and adult CNS. To better understand how Notch signaling is involved in stem cell maintenance during adult neurogenesis in zebrafish we analysed Notch receptor expression in five neurogenic zones of the adult zebrafish brain. Combining proliferation and glial markers we identified several subsets of Notch receptor expressing cells. We found that 90 of proliferating radial glia express notch1a, notch1b and notch3. In contrast, the proliferating non-glial populations of the dorsal telencephalon and hypothalamus rarely express notch3 and about half express notch1a/1b. In the non-proliferating radial glia notch3 is the predominant receptor throughout the brain. In the ventral telencephalon and in the mitotic area of the optic tectum, where cells have neuroepithelial properties, notch1a/1b/3 are expressed in most proliferating cells. However, in the cerebellar niche, although progenitors also have neuroepithelial properties, only notch1a/1b are expressed in a high number of PCNA cells. In this region notch3 expression is mostly in Bergmann glia and at low levels in few PCNA cells. Additionally, we found that in the proliferation zone of the ventral telencephalon, Notch receptors display an apical high to basal low gradient of expression. Notch receptors are also expressed in subpopulations of oligodendrocytes, neurons and endothelial cells. We suggest that the partial regional heterogeneity observed for Notch expression in progenitor cells might be related to the cellular diversity present in each of these neurogenic niches.
Collapse
Affiliation(s)
- Vanessa de Oliveira-Carlos
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Julia Ganz
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Stefan Hans
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jan Kaslin
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
34
|
Zhang X, Huang G, Liu H, Chang H, Wilson JX. Folic acid enhances Notch signaling, hippocampal neurogenesis, and cognitive function in a rat model of cerebral ischemia. Nutr Neurosci 2013; 15:55-61. [DOI: 10.1179/1476830511y.0000000025] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
35
|
The endogenous regenerative capacity of the damaged newborn brain: boosting neurogenesis with mesenchymal stem cell treatment. J Cereb Blood Flow Metab 2013; 33:625-34. [PMID: 23403379 PMCID: PMC3652688 DOI: 10.1038/jcbfm.2013.3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Neurogenesis continues throughout adulthood. The neurogenic capacity of the brain increases after injury by, e.g., hypoxia-ischemia. However, it is well known that in many cases brain damage does not resolve spontaneously, indicating that the endogenous regenerative capacity of the brain is insufficient. Neonatal encephalopathy leads to high mortality rates and long-term neurologic deficits in babies worldwide. Therefore, there is an urgent need to develop more efficient therapeutic strategies. The latest findings indicate that stem cells represent a novel therapeutic possibility to improve outcome in models of neonatal encephalopathy. Transplanted stem cells secrete factors that stimulate and maintain neurogenesis, thereby increasing cell proliferation, neuronal differentiation, and functional integration. Understanding the molecular and cellular mechanisms underlying neurogenesis after an insult is crucial for developing tools to enhance the neurogenic capacity of the brain. The aim of this review is to discuss the endogenous capacity of the neonatal brain to regenerate after a cerebral ischemic insult. We present an overview of the molecular and cellular mechanisms underlying endogenous regenerative processes during development as well as after a cerebral ischemic insult. Furthermore, we will consider the potential to use stem cell transplantation as a means to boost endogenous neurogenesis and restore brain function.
Collapse
|
36
|
Wang Y, Tu W, Lou Y, Xie A, Lai X, Guo F, Deng Z. Mesenchymal stem cells regulate the proliferation and differentiation of neural stem cells through Notch signaling. Cell Biol Int 2013; 33:1173-9. [DOI: 10.1016/j.cellbi.2009.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 05/30/2009] [Accepted: 08/17/2009] [Indexed: 01/22/2023]
|
37
|
Kang SS, Keasey MP, Arnold SA, Reid R, Geralds J, Hagg T. Endogenous CNTF mediates stroke-induced adult CNS neurogenesis in mice. Neurobiol Dis 2012; 49:68-78. [PMID: 22960105 DOI: 10.1016/j.nbd.2012.08.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/16/2012] [Accepted: 08/22/2012] [Indexed: 11/24/2022] Open
Abstract
Focal brain ischemia in adult rats rapidly and robustly induces neurogenesis in the subventricular zone (SVZ) but there are few and inconsistent reports in mice, presenting a hurdle to genetically investigate the endogenous neurogenic regulators such as ciliary neurotrophic factor (CNTF). Here, we first provide a platform for further studies by showing that middle cerebral artery occlusion in adult male C57BL/6 mice robustly enhances neurogenesis in the SVZ only under very specific conditions, i.e., 14days after a 30min occlusion. CNTF expression paralleled changes in the number of proliferated, BrdU-positive, SVZ cells. Stroke-induced proliferation was absent in CNTF-/- mice, suggesting that it is mediated by CNTF. MCAO-increased CNTF appears to act on C cell proliferation and by inducing FGF2 expression but not via EGF expression or Notch1 signaling of neural stem cells in the SVZ. CNTF is unique, as expression of other gp130 ligands, IL-6 and LIF, did not predict SVZ proliferation or showed no or only small compensatory increases in CNTF-/- mice. Expression of tumor necrosis factor-α, which can inhibit neurogenesis, and the presence of leukocytes in the SVZ were inversely correlated with neurogenesis, but pro-inflammatory cytokines did not affect CNTF expression in cultured astrocytes. These results suggest that slowly up-regulated CNTF in the SVZ mediates stroke-induced neurogenesis and is counteracted by inflammation. Further pharmacological stimulation of endogenous CNTF might be a good therapeutic strategy for cell replacement after stroke as CNTF regulates normal patterns of neurogenesis and is expressed almost exclusively in the nervous system.
Collapse
Affiliation(s)
- Seong Su Kang
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Matthew P Keasey
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Sheila A Arnold
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA; Department of Pharmacology and Toxicology, University of Louisville, KY 40292, USA
| | - Rollie Reid
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Justin Geralds
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Theo Hagg
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA; Department of Pharmacology and Toxicology, University of Louisville, KY 40292, USA.
| |
Collapse
|
38
|
Mishra M, Heese K. P60TRP interferes with the GPCR/secretase pathway to mediate neuronal survival and synaptogenesis. J Cell Mol Med 2012; 15:2462-77. [PMID: 21199326 PMCID: PMC3822957 DOI: 10.1111/j.1582-4934.2010.01248.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
In the present study, we show that overexpression of the G-protein-coupled receptor (GPCR)-associated sorting protein p60TRP (transcription regulator protein) in neural stem cells (NSCs) and in a transgenic mouse model modulates the phosphorylation and proteolytic processing of amyloid precursor protein (App), N-cadherin (Cdh2), presenilin (Psen) and τ protein (Mapt). Our results suggest that p60TRP is an inhibitor of Bace1 (β-site App cleaving enzyme) and Psen. We performed several apoptosis assays [Annexin-V, TdT-mediated dUTP Nick-End Labeling (TUNEL), caspase-3/7] using NSCs and PC12 cells (overexpressing p60TRP and knockdown of p60TRP) to substantiate the neuroprotective role of p60TRP. Functional analyses, both in vitro and in vivo, revealed that p60TRP promotes neurosynaptogenesis. Characterization of the cognitive function of p60TRP transgenic mice using the radial arm water maze test demonstrated that p60TRP improved memory and learning abilities. The improved cognitive functions could be attributed to increased synaptic connections and plasticity, which was confirmed by the modulation of the γ-aminobutyric acid receptor system and the elevated expression of microtubule-associated protein 2, synaptophysin and Slc17a7 (vesicle glutamate transporter, Vglut1), as well as by the inhibition of Cdh2 cleavage. In conclusion, interference with the p60TRP/ GPCR/secretase signalling pathway might be a new therapeutic target for the treatment of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Manisha Mishra
- Department of Molecular and Cell Biology, School of Biological Sciences, College of Science, Nanyang Technological University, Singapore
| | | |
Collapse
|
39
|
Stessin AM, Gursel DB, Schwartz A, Parashar B, Kulidzhanov FG, Sabbas AM, Boockvar J, Nori D, Wernicke AG. FTY720, sphingosine 1-phosphate receptor modulator, selectively radioprotects hippocampal neural stem cells. Neurosci Lett 2012; 516:253-8. [PMID: 22507238 DOI: 10.1016/j.neulet.2012.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/30/2012] [Accepted: 04/01/2012] [Indexed: 12/27/2022]
Abstract
Cranial irradiation is an effective treatment modality for both primary and metastatic brain tumors, yet it induces cognitive decline in a substantial number of patients. At present, there are no established methods for neuroprotection. Recent investigations have revealed a link between radiation-induced cognitive dysfunction and the loss of neural precursor cells in the hippocampus. Hence, identifying pharmacological agents, capable of protecting this cell population, is of interest. FTY720 (fingolimod), an FDA-approved oral drug for the treatment of multiple sclerosis, has been shown to promote the survival and differentiation of neural progenitors, as well as remyelination and repair after brain injury. In this study, we show that FTY720, used at nanomolar concentrations, is capable of increasing the viability and neurogenicity of irradiated neural stem cells from the hippocampus. In contrast, it does not provide radioprotection in a human breast cancer cell line and two glioma cell lines. These results suggest a potential therapeutic role for FTY720 as a neuroprotector during cranial irradiation. Further preclinical studies are warranted to evaluate this possibility.
Collapse
Affiliation(s)
- Alexander M Stessin
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ohta S, Misawa A, Fukaya R, Inoue S, Kanemura Y, Okano H, Kawakami Y, Toda M. Macrophage migration inhibitory factor (MIF) promotes cell survival and proliferation of neural stem/progenitor cells. J Cell Sci 2012; 125:3210-20. [PMID: 22454509 DOI: 10.1242/jcs.102210] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In a previous study, we showed that murine dendritic cells (DCs) can increase the number of neural stem/progenitor cells (NSPCs) in vitro and in vivo. In the present study, we identified macrophage migration inhibitory factor (MIF) as a novel factor that can support the proliferation and/or survival of NSPCs in vitro. MIF is secreted by DCs and NSPCs, and its function in the normal brain remains largely unknown. It was previously shown that in macrophages, MIF binds to a CD74-CD44 complex. In the present study, we observed the expression of MIF receptors in mouse ganglionic-eminence-derived neurospheres using flow cytometry in vitro. We also found CD74 expression in the ganglionic eminence of E14 mouse brains, suggesting that MIF plays a physiological role in vivo. MIF increased the number of primary and secondary neurospheres. By contrast, retrovirally expressed MIF shRNA and MIF inhibitor (ISO-1) suppressed primary and secondary neurosphere formation, as well as cell proliferation. In the neurospheres, MIF knockdown by shRNA increased caspase 3/7 activity, and MIF increased the phosphorylation of Akt, Erk, AMPK and Stat3 (Ser727), as well as expression of Hes3 and Egfr, the products of which are known to support cell survival, proliferation and/or maintenance of NSPCs. MIF also acted as a chemoattractant for NSPCs. These results show that MIF can induce NSPC proliferation and maintenance by multiple signaling pathways acting synergistically, and it may be a potential therapeutic factor, capable of activating NSPC, for the treatment of degenerative brain disorders.
Collapse
Affiliation(s)
- Shigeki Ohta
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cytotoxic T lymphocyte antigen 4 immunogloblin promotes neuronal differentiation in the grafts of embryonic stem cell-derived neural precursor cells. Neuroscience 2012; 202:484-91. [DOI: 10.1016/j.neuroscience.2011.11.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Revised: 11/09/2011] [Accepted: 11/24/2011] [Indexed: 12/31/2022]
|
42
|
Covey MV, Loporchio D, Buono KD, Levison SW. Opposite effect of inflammation on subventricular zone versus hippocampal precursors in brain injury. Ann Neurol 2011; 70:616-26. [PMID: 21710624 PMCID: PMC3522429 DOI: 10.1002/ana.22473] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 04/14/2011] [Accepted: 04/22/2011] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Inflammation promotes epidermal wound healing but is considered detrimental to recovery from central nervous system injury. Sick infants have increased levels of cytokines in their cerebrospinal fluid that correlate with poor neurological outcome. In this study, we investigated the role of neuroinflammation and more specifically interleukin 6 (IL-6) in the amplification of subventricular zone (SVZ) and subgranular zone (SGZ) neural precursors after neonatal brain injury. METHODS Neonatal hypoxia/ischemia (H/I) was induced in P6 rat pups, and IL-6 was quantified with or without indomethacin administration. Neural precursor responses were evaluated by neurosphere assays as well as by stereological analyses. Studies were performed to determine how IL-6 and leukemia-inhibiting factor (LIF) affect SVZ cell expansion, proliferation, and self-renewal. RESULTS Consistent with earlier studies, medially situated SVZ cells expanded after H/I. Contrary to our expectations, indomethacin significantly decreased both the initial reactive increase in these precursors and their ability to self-renew. By contrast, indomethacin increased proliferation in the SGZ and lateral SVZ. Indomethacin diminished the accumulation of microglia/macrophages and IL-6 production after H/I. In vitro IL-6 enhanced neurosphere growth, self-renewal, and tripotentiality and was more effective than LIF in promoting self-renewal. Enhanced precursor self-renewal also was obtained using prostaglandin E2, which is downstream of cyclooxygenase 2 and a target of indomethacin. INTERPRETATION These data implicate neuroinflammation and in particular IL-6 as a positive effector of primitive neural precursor expansion after neonatal brain injury. These findings have important clinical implications, as indomethacin and other anti-inflammatory agents are administered to premature infants for a variety of reasons.
Collapse
Affiliation(s)
- Matthew V Covey
- Department of Neurology and Neurosciences, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, 205 South Orange Avenue, Newark, NJ 07103, USA
| | | | | | | |
Collapse
|
43
|
Pauklin M, Thomasen H, Pester A, Steuhl KP, Meller D. Expression of Pluripotency and Multipotency Factors in Human Ocular Surface Tissues. Curr Eye Res 2011; 36:1086-97. [DOI: 10.3109/02713683.2011.608238] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mikk Pauklin
- Department of Ophthalmology, University of Duisburg-Essen,
Essen, Germany
- Eye Clinic, Tartu University Hospital,
Tartu, Estonia
| | - Henning Thomasen
- Department of Ophthalmology, University of Duisburg-Essen,
Essen, Germany
| | - Anne Pester
- Department of Ophthalmology, University of Duisburg-Essen,
Essen, Germany
| | - Klaus-P. Steuhl
- Department of Ophthalmology, University of Duisburg-Essen,
Essen, Germany
| | - Daniel Meller
- Department of Ophthalmology, University of Duisburg-Essen,
Essen, Germany
| |
Collapse
|
44
|
Russo I, Amornphimoltham P, Weigert R, Barlati S, Bosetti F. Cyclooxygenase-1 is involved in the inhibition of hippocampal neurogenesis after lipopolysaccharide-induced neuroinflammation. Cell Cycle 2011; 10:2568-73. [PMID: 21694498 PMCID: PMC3180195 DOI: 10.4161/cc.10.15.15946] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 05/26/2011] [Accepted: 05/31/2011] [Indexed: 12/21/2022] Open
Abstract
Growing evidence indicates that neuroinflammation can alter adult neurogenesis by mechanisms as yet unclear. We have previously demonstrated that the neuroinflammatory response and neuronal damage after lipopolysaccharide (LPS) injection is reduced in cyclooxygenase-1 deficient (COX-1(-/-)) mice. In this study, we investigated the role of COX-1 on hippocampal neurogenesis during LPS-induced neuroinflammation, using COX-1(-/-) and wild type (WT) mice. We found that LPS-induced neuroinflammation resulted in the decrease of proliferation, survival and differentiation of hippocampal progenitor cells in WT but not in COX-1(-/-) mice. Thus, we demonstrate for the first time that COX-1 is involved in the inhibition of BrdU progenitor cells in proliferation and hippocampal neurogenesis after LPS. These results suggest that COX-1 may represent a viable therapeutic target to reduce neuroinflammation and promote neurogenesis in neurodegenerative diseases with a strong inflammatory component.
Collapse
Affiliation(s)
- Isabella Russo
- Molecular Neuroscience Unit; Brain Physiology and Metabolism Section; National Institute on Aging; National Institutes of Health; Bethesda, MD USA
- Division of Biology and Genetics; Department of Biomedical Sciences and Biotechnologies and National Institute of Neuroscience; University of Brescia; Brescia, Italy
| | - Panomwat Amornphimoltham
- Intracellular Membrane Trafficking Unit; Oral and Pharyngeal Cancer Branch; National Institute of Dental and Craniofacial Research; National Institutes of Health; Bethesda, MD USA
| | - Roberto Weigert
- Intracellular Membrane Trafficking Unit; Oral and Pharyngeal Cancer Branch; National Institute of Dental and Craniofacial Research; National Institutes of Health; Bethesda, MD USA
| | - Sergio Barlati
- Division of Biology and Genetics; Department of Biomedical Sciences and Biotechnologies and National Institute of Neuroscience; University of Brescia; Brescia, Italy
| | - Francesca Bosetti
- Molecular Neuroscience Unit; Brain Physiology and Metabolism Section; National Institute on Aging; National Institutes of Health; Bethesda, MD USA
| |
Collapse
|
45
|
Starossom SC, Imitola J, Wang Y, Cao L, Khoury SJ. Subventricular zone microglia transcriptional networks. Brain Behav Immun 2011; 25:991-9. [PMID: 21074605 PMCID: PMC3109092 DOI: 10.1016/j.bbi.2010.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 11/03/2010] [Accepted: 11/03/2010] [Indexed: 01/19/2023] Open
Abstract
Microglia play an important role in inflammatory diseases of the central nervous system. There is evidence of microglial diversity with distinct phenotypes exhibiting either neuroprotection and repair or neurotoxicity. However the precise molecular mechanisms underlying this diversity are still unknown. Using a model of experimental autoimmune encephalomyelitis (EAE) we performed transcriptional profiling of isolated subventricular zone microglia from the acute and chronic disease phases of EAE. We found that microglia exhibit disease phase specific gene expression signatures, that correspond to unique gene ontology functions and genomic networks. Our data demonstrate for the first time, distinct transcriptional networks of microglia activation in vivo, that suggests a role as mediators of injury or repair.
Collapse
Affiliation(s)
- Sarah C. Starossom
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Jaime Imitola
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Yue Wang
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Li Cao
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Samia J. Khoury
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
46
|
Abstract
The Notch pathway is prominent among those known to regulate neural development in vertebrates. Notch receptor activation can inhibit neurogenesis, maintain neural progenitor character, and in some contexts promote gliogenesis and drive binary fate choices. Recently, a wave of exciting studies has emerged, which has both solidified previously held assertions and expanded our understanding of Notch function during neurogenesis and in the adult brain. These studies have examined pathway regulators and interactions, as well as pathway dynamics, with respect to both gene expression and cell-cell signaling. Here, focusing primarily on vertebrates, we review the current literature on Notch signaling in the nervous system, and highlight numerous recent studies that have generated interesting and unexpected advances.
Collapse
|
47
|
Gomi M, Aoki T, Takagi Y, Nishimura M, Ohsugi Y, Mihara M, Nozaki K, Hashimoto N, Miyamoto S, Takahashi J. Single and local blockade of interleukin-6 signaling promotes neuronal differentiation from transplanted embryonic stem cell-derived neural precursor cells. J Neurosci Res 2011; 89:1388-99. [PMID: 21557295 DOI: 10.1002/jnr.22667] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 03/17/2011] [Accepted: 03/24/2011] [Indexed: 11/08/2022]
Abstract
Safe and efficient transplantation of embryonic stem (ES) cells to the brain requires that local inflammatory and immune responses to allogeneic grafts are inhibited. To investigate cytokines that affect graft cell survival and differentiation, we used stromal cell-derived inducing activity to induce the differentiation of neural progenitor cells (NPCs) from mouse ES cells and transplanted the NPCs into mouse brain. Examination of surrounding brain tissue revealed elevated expression levels of interleukin (IL)-1β, IL-4, and IL-6 in response to NPC transplantation. Among these, only IL-6 reduced neuronal differentiation and promoted glial differentiation in vitro. When we added anti-IL-6 receptor antibodies to NPCs during transplantation, this single and local blockade of IL-6 signaling reduced the accumulation of host-derived leukocytes, including microglia. Furthermore, it also promoted neuronal differentiation and reduced glial differentiation from the grafted NPCs to an extent similar to that with systemic and continuous administration of cyclosporine A. These results suggest that local administration of anti-IL-6 receptor antibodies with NPCs may promote neuronal differentiation during the treatment of neurological diseases with cell replacement therapy.
Collapse
Affiliation(s)
- Masanori Gomi
- Department of Biological Repair, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wianny F, Bourillot PY, Dehay C. Embryonic stem cells in non-human primates: An overview of neural differentiation potential. Differentiation 2011; 81:142-52. [PMID: 21296479 DOI: 10.1016/j.diff.2011.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/18/2010] [Accepted: 01/11/2011] [Indexed: 12/11/2022]
Abstract
Non-human primate (NHP) embryonic stem (ES) cells show unlimited proliferative capacities and a great potential to generate multiple cell lineages. These properties make them an ideal resource both for investigating early developmental processes and for assessing their therapeutic potential in numerous models of degenerative diseases. They share the same markers and the same properties with human ES cells, and thus provide an invaluable transitional model that can be used to address the safety issues related to the clinical use of human ES cells. Here, we review the available information on the derivation and the specific features of monkey ES cells. We comment on the capacity of primate ES cells to differentiate into neural lineages and the current protocols to generate self-renewing neural stem cells. We also highlight the signalling pathways involved in the maintenance of these neural cell types. Finally, we discuss the potential of monkey ES cells for neuronal differentiation.
Collapse
Affiliation(s)
- Florence Wianny
- Inserm, U846, Stem Cell and Brain Research Institute, 18 Avenue Doyen Lépine, 69500 Bron, France.
| | | | | |
Collapse
|
49
|
Prospects and limitations of using endogenous neural stem cells for brain regeneration. Genes (Basel) 2011; 2:107-30. [PMID: 24710140 PMCID: PMC3924842 DOI: 10.3390/genes2010107] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 12/06/2010] [Accepted: 01/04/2011] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells (NSCs) are capable of producing a variety of neural cell types, and are indispensable for the development of the mammalian brain. NSCs can be induced in vitro from pluripotent stem cells, including embryonic stem cells and induced-pluripotent stem cells. Although the transplantation of these exogenous NSCs is a potential strategy for improving presently untreatable neurological conditions, there are several obstacles to its implementation, including tumorigenic, immunological, and ethical problems. Recent studies have revealed that NSCs also reside in the adult brain. The endogenous NSCs are activated in response to disease or trauma, and produce new neurons and glia, suggesting they have the potential to regenerate damaged brain tissue while avoiding the above-mentioned problems. Here we present an overview of the possibility and limitations of using endogenous NSCs in regenerative medicine.
Collapse
|
50
|
Zhou ZD, Kumari U, Xiao ZC, Tan EK. Notch as a molecular switch in neural stem cells. IUBMB Life 2010; 62:618-23. [PMID: 20681026 DOI: 10.1002/iub.362] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Notch signaling pathway, originally discovered in Drosophila, impinges on a wide array of cellular processes including cell proliferation, cell differentiation, and cell apoptosis. Recent accumulating evidences implicated the important roles of Notch signal pathway in different aspects of stem cell biology of neural stem cell (NSC). In vivo and in vitro studies illustrated that Notch signal pathway could promote gliogenesis, inhibit premature neurogenesis, and be involved in self-renew of NSC. This short review summarizes the roles of the Notch signaling pathway on gliogenesis, neurogenesis, and self-renew of NSC and their underlying molecular mechanisms.
Collapse
Affiliation(s)
- Zhi-Dong Zhou
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore
| | | | | | | |
Collapse
|