1
|
Shen W, Jiang N, Zhou W. What can traditional Chinese medicine do for adult neurogenesis? Front Neurosci 2023; 17:1158228. [PMID: 37123359 PMCID: PMC10130459 DOI: 10.3389/fnins.2023.1158228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/13/2023] [Indexed: 05/02/2023] Open
Abstract
Adult neurogenesis plays a crucial role in cognitive function and mood regulation, while aberrant adult neurogenesis contributes to various neurological and psychiatric diseases. With a better understanding of the significance of adult neurogenesis, the demand for improving adult neurogenesis is increasing. More and more research has shown that traditional Chinese medicine (TCM), including TCM prescriptions (TCMPs), Chinese herbal medicine, and bioactive components, has unique advantages in treating neurological and psychiatric diseases by regulating adult neurogenesis at various stages, including proliferation, differentiation, and maturation. In this review, we summarize the progress of TCM in improving adult neurogenesis and the key possible mechanisms by which TCM may benefit it. Finally, we suggest the possible strategies of TCM to improve adult neurogenesis in the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wei Shen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Ning Jiang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- *Correspondence: Ning Jiang, ; Wenxia Zhou,
| | - Wenxia Zhou
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- *Correspondence: Ning Jiang, ; Wenxia Zhou,
| |
Collapse
|
2
|
Chai Y, Zhao H, Yang S, Gao X, Cao Z, Lu J, Sun Q, Liu W, Zhang Z, Yang J, Wang X, Chen T, Kong X, Mikos AG, Zhang X, Zhang Y, Wang X. Structural alignment guides oriented migration and differentiation of endogenous neural stem cells for neurogenesis in brain injury treatment. Biomaterials 2021; 280:121310. [PMID: 34890972 DOI: 10.1016/j.biomaterials.2021.121310] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023]
Abstract
Radial glia (RG) cells that align in parallel in the embryonic brain are found to be able to guide the directed migration of neurons in response to brain injury. Therefore, biomaterials with aligned architectures are supposed to have positive effects on neural migration and neurogenic differentiation for brain injury repair that are rarely addressed, although they have been widely demonstrated in spinal cord and peripheral nerve system. Here, we present a highly biomimetic scaffold of aligned fibrin hydrogel (AFG) that mimics the oriented structure of RG fibers. Through a combination of histological, behavioral, imaging, and transcriptomic analyses, we demonstrated that transplanting the AFG scaffold into injured cortical brains promotes effective migration, differentiation, and maturation of endogenous neural stem cells, resulting in neurological functional recovery. Therefore, this study will light up a new perspective on applying an aligned scaffold to promote cortical regeneration after injury by inducing endogenous neurogenesis.
Collapse
Affiliation(s)
- Yi Chai
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China; Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China; Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160, Pujian Road, District Pudong, Shanghai, 200127, China
| | - He Zhao
- Department of orthopacdic III, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Xiaohan Gao
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China
| | - Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Jiaju Lu
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qingling Sun
- Department of orthopacdic III, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Wei Liu
- Department of Neurosurgery, The First Hospital of Hebei Medical University, Hebei, 050000, China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Junyi Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Xuelin Wang
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, China
| | - Tuoyu Chen
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China
| | - Xiangdong Kong
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Xiaohua Zhang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160, Pujian Road, District Pudong, Shanghai, 200127, China
| | - Yuqi Zhang
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China.
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
3
|
Brown C, McKee C, Halassy S, Kojan S, Feinstein DL, Chaudhry GR. Neural stem cells derived from primitive mesenchymal stem cells reversed disease symptoms and promoted neurogenesis in an experimental autoimmune encephalomyelitis mouse model of multiple sclerosis. Stem Cell Res Ther 2021; 12:499. [PMID: 34503569 PMCID: PMC8427882 DOI: 10.1186/s13287-021-02563-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system (CNS). MS affects millions of people and causes a great economic and societal burden. There is no cure for MS. We used a novel approach to investigate the therapeutic potential of neural stem cells (NSCs) derived from human primitive mesenchymal stem cells (MSCs) in an experimental autoimmune encephalomyelitis (EAE) mouse model of MS. Methods MSCs were differentiated into NSCs, labeled with PKH26, and injected into the tail vein of EAE mice. Neurobehavioral changes in the mice assessed the effect of transplanted cells on the disease process. The animals were sacrificed two weeks following cell transplantation to collect blood, lymphatic, and CNS tissues for analysis. Transplanted cells were tracked in various tissues by flow cytometry. Immune infiltrates were determined and characterized by H&E and immunohistochemical staining, respectively. Levels of immune regulatory cells, Treg and Th17, were analyzed by flow cytometry. Myelination was determined by Luxol fast blue staining and immunostaining. In vivo fate of transplanted cells and expression of inflammation, astrogliosis, myelination, neural, neuroprotection, and neurogenesis markers were investigated by using immunohistochemical and qRT-PCR analysis.
Results MSC-derived NSCs expressed specific neural markers, NESTIN, TUJ1, VIMENTIN, and PAX6. NSCs improved EAE symptoms more than MSCs when transplanted in EAE mice. Post-transplantation analyses also showed homing of MSCs and NSCs into the CNS with concomitant induction of an anti-inflammatory response, resulting in reducing immune infiltrates. NSCs also modulated Treg and Th17 cell levels in EAE mice comparable to healthy controls. Luxol fast blue staining showed significant improvement in myelination in treated mice. Further analysis showed that NSCs upregulated genes involved in myelination and neuroprotection but downregulated inflammatory and astrogliosis genes more significantly than MSCs. Importantly, NSCs differentiated into neural derivatives and promoted neurogenesis, possibly by modulating BDNF and FGF signaling pathways. Conclusions NSC transplantation reversed the disease process by inducing an anti-inflammatory response and promoting myelination, neuroprotection, and neurogenesis in EAE disease animals. These promising results provide a basis for clinical studies to treat MS using NSCs derived from primitive MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02563-8.
Collapse
Affiliation(s)
- Christina Brown
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Christina McKee
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Sophia Halassy
- Ascension Providence Hospital, Southfield, MI, 48075, USA
| | - Suleiman Kojan
- Department of Neuroscience, OUWB School of Medicine, Oakland University, Rochester, MI, 48309, USA
| | - Doug L Feinstein
- Department of Anesthesiology, The University of Illinois at Chicago, Chicago, IL, 60607, USA.,Department of Veterans Affairs, Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA.
| |
Collapse
|
4
|
Hassn Mesrati M, Behrooz AB, Y. Abuhamad A, Syahir A. Understanding Glioblastoma Biomarkers: Knocking a Mountain with a Hammer. Cells 2020; 9:E1236. [PMID: 32429463 PMCID: PMC7291262 DOI: 10.3390/cells9051236] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
Gliomas are the most frequent and deadly form of human primary brain tumors. Among them, the most common and aggressive type is the high-grade glioblastoma multiforme (GBM), which rapidly grows and renders patients a very poor prognosis. Meanwhile, cancer stem cells (CSCs) have been determined in gliomas and play vital roles in driving tumor growth due to their competency in self-renewal and proliferation. Studies of gliomas have recognized CSCs via specific markers. This review comprehensively examines the current knowledge of the most significant CSCs markers in gliomas in general and in glioblastoma in particular and specifically focuses on their outlook and importance in gliomas CSCs research. We suggest that CSCs should be the superior therapeutic approach by directly targeting the markers. In addition, we highlight the association of these markers with each other in relation to their cascading pathways, and interactions with functional miRNAs, providing the role of the networks axes in glioblastoma signaling pathways.
Collapse
Affiliation(s)
| | | | | | - Amir Syahir
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia; (M.H.M.); (A.B.B.); (A.Y.A.)
| |
Collapse
|
5
|
Purvis EM, O'Donnell JC, Chen HI, Cullen DK. Tissue Engineering and Biomaterial Strategies to Elicit Endogenous Neuronal Replacement in the Brain. Front Neurol 2020; 11:344. [PMID: 32411087 PMCID: PMC7199479 DOI: 10.3389/fneur.2020.00344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
Neurogenesis in the postnatal mammalian brain is known to occur in the dentate gyrus of the hippocampus and the subventricular zone. These neurogenic niches serve as endogenous sources of neural precursor cells that could potentially replace neurons that have been lost or damaged throughout the brain. As an example, manipulation of the subventricular zone to augment neurogenesis has become a popular strategy for attempting to replace neurons that have been lost due to acute brain injury or neurodegenerative disease. In this review article, we describe current experimental strategies to enhance the regenerative potential of endogenous neural precursor cell sources by enhancing cell proliferation in neurogenic regions and/or redirecting migration, including pharmacological, biomaterial, and tissue engineering strategies. In particular, we discuss a novel replacement strategy based on exogenously biofabricated "living scaffolds" that could enhance and redirect endogenous neuroblast migration from the subventricular zone to specified regions throughout the brain. This approach utilizes the first implantable, biomimetic tissue-engineered rostral migratory stream, thereby leveraging the brain's natural mechanism for sustained neuronal replacement by replicating the structure and function of the native rostral migratory stream. Across all these strategies, we discuss several challenges that need to be overcome to successfully harness endogenous neural precursor cells to promote nervous system repair and functional restoration. With further development, the diverse and innovative tissue engineering and biomaterial strategies explored in this review have the potential to facilitate functional neuronal replacement to mitigate neurological and psychiatric symptoms caused by injury, developmental disorders, or neurodegenerative disease.
Collapse
Affiliation(s)
- Erin M. Purvis
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - John C. O'Donnell
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - H. Isaac Chen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
6
|
Yang Q, Wu J, Zhao J, Xu T, Han P, Song X. The Expression Profiles of lncRNAs and Their Regulatory Network During Smek1/2 Knockout Mouse Neural Stem Cells Differentiation. Curr Bioinform 2020. [DOI: 10.2174/1574893614666190308160507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background:
Previous studies indicated that the cell fate of neural stem cells (NSCs)
after differentiation is determined by Smek1, one isoform of suppressor of Mek null (Smek). Smek
deficiency prevents NSCs from differentiation, thus affects the development of nervous system. In
recent years, lncRNAs have been found to participate in numerous developmental and biological
pathways. However, the effects of knocking out Smek on the expression profiles of lncRNAs
during the differentiation remain unknown.
Objective:
This study is to explore the expression profiles of lncRNAs and their possible function
during the differentiation from Smek1/2 knockout NSCs.
Methods:
We obtained NSCs from the C57BL/6J mouse fetal cerebral cortex. One group of NSCs
was from wildtype mouse (WT group), while another group was from knocked out Smek1/2 (KO
group).
Results:
By analyzing the RNA-Seq data, we found that after knocking out Smek1/2, the
expression profiles of mRNAs and lncRNAs revealed significant changes. Analyses indicated that
these affected mRNAs have connections with the pathway network for the differentiation and
proliferation of NSCs. Furthermore, we performed a co-expression network analysis on the
differentially expressed mRNAs and lncRNAs, which helped reveal the possible regulatory rules
of lncRNAs during the differentiation after knocking out Smek1/2.
Conclusion:
By comparing group WT with KO, we found 366 differentially expressed mRNAs
and 12 lncRNAs. GO and KEGG enrichment analysis on these mRNAs suggested their
relationships with differentiation and proliferation of NSCs. Some of these mRNAs and lncRNAs
have been verified to play regulatory roles in nervous system. Analyses on the co-expression
network also indicated the possible functions of affected mRNAs and lncRNAs during NSCs
differentiation after knocking out Smek1/2.
Collapse
Affiliation(s)
- Qichang Yang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Jing Wu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Jian Zhao
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Tianyi Xu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Ping Han
- The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210019, China
| | - Xiaofeng Song
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| |
Collapse
|
7
|
Yang C, Zhang X, Yin H, Du Z, Yang Z. MiR-429/200a/200b negatively regulate Notch1 signaling pathway to suppress CoCl 2-induced apoptosis in PC12 cells. Toxicol In Vitro 2020; 65:104787. [PMID: 32004541 DOI: 10.1016/j.tiv.2020.104787] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/11/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
Abstract
Neuronal apoptosis is a central hallmark of cerebral ischemia, which is serious threats to human health. Notch1 signaling pathway and three members of miR-200 family, miR-429, miR-200a and miR-200b, are reported to have tight connection with hypoxia-induced injury. However, their mutual regulation relationship and their roles in neuronal apoptosis caused by hypoxia are rarely reported. In the present study, differentiated pheochromocytoma (PC12) cells were treated with chemical hypoxia inducer, cobalt chloride (CoCl2) to establish in vitro neuronal hypoxia model. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, Western blot assay and Hoechst staining indicated that CoCl2 caused apoptosis of PC12 cells along with the activation of Notch1 signallilng pathway. The treatment of N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butylester (DAPT) inhibited Notch1 signaling pathway and attenuated the apoptosis induced by CoCl2. Real-time polymerase chain reaction (RT-PCR) showed that expressions of miR-429/200a/200b were dynamically changed during the treatment of CoCl2, and significantly decreased after 12-hour treatment of CoCl2. Overexpression of miR-429/200a/200b inhibited the Notch1 signaling pathway and suppressed CoCl2-induced apoptosis in PC12 cells. These results may clarify the roles of miR-429/200a/200b and Notch1 signaling pathway in hypoxia-induced nerve injury and provide a new theoretical basis to relieve nerve injury.
Collapse
Affiliation(s)
- Chunxiao Yang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Xiaochen Zhang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Hongqiang Yin
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Zhanqiang Du
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Zhuo Yang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
8
|
Sarubbo F, Moranta D, Pani G. Dietary polyphenols and neurogenesis: Molecular interactions and implication for brain ageing and cognition. Neurosci Biobehav Rev 2018; 90:456-470. [DOI: 10.1016/j.neubiorev.2018.05.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 04/05/2018] [Accepted: 05/07/2018] [Indexed: 12/17/2022]
|
9
|
Li Q, Zhang X, Cheng N, Yang C, Zhang T. Notch1 knockdown disturbed neural oscillations in the hippocampus of C57BL mice. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:63-70. [PMID: 29410010 DOI: 10.1016/j.pnpbp.2018.01.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 01/17/2018] [Accepted: 01/29/2018] [Indexed: 12/30/2022]
Abstract
Neural oscillations and their interactions are associated with the coordination of neural groups, which provide a mechanism underlying information processing of brain functions. Notch1 receptor is involved in the neurological and psychiatric disorders, such as neurodevelopmental deficits, cerebral ischemia, Alzheimer's disease and depression. Here, we investigated the dynamics of neural oscillations in hippocampus of Notch1+/- mice in urethane-anesthetized state. Notch1 knockdown altered the distribution of power in the hippocampal DG areas, reduced theta (3-8 Hz) power and enhanced low gamma (LG, 30-50 Hz) and high gamma (HG, 50-100 Hz) power. Moreover, theta-gamma phase-amplitude coupling in the hippocampal DG area was markedly impaired in the Notch1+/- mice. The data further showed that the expression of NR2B was decreased, and the expressions of GABAARα1, GAD67 and parvalbumin were considerably increased after Notch1 knockdown. Taken together, our results suggest that Notch1 genetic deficiency significantly impaired the corss-frequency coupling of neural oscillations, and their interactions in the hippocampal DG region by means of disrupting the balance of excitatory and inhibitory receptors, which could be an underlying mechanism of cognitive impairment in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Qun Li
- College of Life Sciences, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, 300071 Tianjin, PR China
| | - Xiaochen Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 300071 Tianjin, PR China
| | - Ning Cheng
- College of Life Sciences, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, 300071 Tianjin, PR China
| | - Chunxiao Yang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 300071 Tianjin, PR China
| | - Tao Zhang
- College of Life Sciences, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, 300071 Tianjin, PR China.
| |
Collapse
|
10
|
O'Léime CS, Cryan JF, Nolan YM. Nuclear deterrents: Intrinsic regulators of IL-1β-induced effects on hippocampal neurogenesis. Brain Behav Immun 2017; 66:394-412. [PMID: 28751020 DOI: 10.1016/j.bbi.2017.07.153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/15/2017] [Accepted: 07/23/2017] [Indexed: 12/11/2022] Open
Abstract
Hippocampal neurogenesis, the process by which new neurons are born and develop into the host circuitry, begins during embryonic development and persists throughout adulthood. Over the last decade considerable insights have been made into the role of hippocampal neurogenesis in cognitive function and the cellular mechanisms behind this process. Additionally, an increasing amount of evidence exists on the impact of environmental factors, such as stress and neuroinflammation on hippocampal neurogenesis and subsequent impairments in cognition. Elevated expression of the pro-inflammatory cytokine interleukin-1β (IL-1β) in the hippocampus is established as a significant contributor to the neuronal demise evident in many neurological and psychiatric disorders and is now known to negatively regulate hippocampal neurogenesis. In order to prevent the deleterious effects of IL-1β on neurogenesis it is necessary to identify signalling pathways and regulators of neurogenesis within neural progenitor cells that can interact with IL-1β. Nuclear receptors are ligand regulated transcription factors that are involved in modulating a large number of cellular processes including neurogenesis. In this review we focus on the signalling mechanisms of specific nuclear receptors involved in regulating neurogenesis (glucocorticoid receptors, peroxisome proliferator activated receptors, estrogen receptors, and nuclear receptor subfamily 2 group E member 1 (NR2E1 or TLX)). We propose that these nuclear receptors could be targeted to inhibit neuroinflammatory signalling pathways associated with IL-1β. We discuss their potential to be therapeutic targets for neuroinflammatory disorders affecting hippocampal neurogenesis and associated cognitive function.
Collapse
Affiliation(s)
- Ciarán S O'Léime
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
11
|
Voluntary running-enhanced synaptic plasticity, learning and memory are mediated by Notch1 signal pathway in C57BL mice. Brain Struct Funct 2017; 223:749-767. [DOI: 10.1007/s00429-017-1521-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 09/04/2017] [Indexed: 12/19/2022]
|
12
|
Kawaguchi-Niida M, Shibata N, Furuta Y. Smad4 is essential for directional progression from committed neural progenitor cells through neuronal differentiation in the postnatal mouse brain. Mol Cell Neurosci 2017; 83:55-64. [PMID: 28669622 DOI: 10.1016/j.mcn.2017.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/01/2017] [Accepted: 06/25/2017] [Indexed: 01/19/2023] Open
Abstract
Signaling by the TGFβ super-family, consisting of TGFβ/activin- and bone morphogenetic protein (BMP) branch pathways, is involved in the central nervous system patterning, growth, and differentiation during embryogenesis. Neural progenitor cells are implicated in various pathological conditions, such as brain injury, infarction, Parkinson's disease and Alzheimer's disease. However, the roles of TGFβ/BMP signaling in the postnatal neural progenitor cells in the brain are still poorly understood. We examined the functional contribution of Smad4, a key integrator of TGFβ/BMP signaling pathways, to the regulation of neural progenitor cells in the subventricular zone (SVZ). Conditional loss of Smad4 in neural progenitor cells caused an increase in the number of neural stem like cells in the SVZ. Smad4 conditional mutants also exhibited attenuation in neuronal lineage differentiation in the adult brain that led to a deficit in olfactory bulb neurons as well as to a reduction of brain parenchymal volume. SVZ-derived neural stem/progenitor cells from the Smad4 mutant brains yielded increased growth of neurospheres, elevated self-renewal capacity and resistance to differentiation. These results indicate that loss of Smad4 in neural progenitor cells causes defects in progression of neural progenitor cell commitment within the SVZ and subsequent neuronal differentiation in the postnatal mouse brain.
Collapse
Affiliation(s)
- Motoko Kawaguchi-Niida
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan; Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Noriyuki Shibata
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit and Genetic Engineering Team, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe, Japan; Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Moon BS, Yun HM, Chang WH, Steele BH, Cai M, Choi SH, Lu W. Smek promotes corticogenesis through regulating Mbd3's stability and Mbd3/NuRD complex recruitment to genes associated with neurogenesis. PLoS Biol 2017; 15:e2001220. [PMID: 28467410 PMCID: PMC5414985 DOI: 10.1371/journal.pbio.2001220] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/06/2017] [Indexed: 11/18/2022] Open
Abstract
The fate of neural progenitor cells (NPCs) during corticogenesis is determined by a complex interplay of genetic or epigenetic components, but the underlying mechanism is incompletely understood. Here, we demonstrate that Suppressor of Mek null (Smek) interact with methyl-CpG-binding domain 3 (Mbd3) and the complex plays a critical role in self-renewal and neuronal differentiation of NPCs. We found that Smek promotes Mbd3 polyubiquitylation and degradation, blocking recruitment of the repressive Mbd3/nucleosome remodeling and deacetylase (NuRD) complex at the neurogenesis-associated gene loci, and, as a consequence, increasing acetyl histone H3 activity and cortical neurogenesis. Furthermore, overexpression of Mbd3 significantly blocked neuronal differentiation of NPCs, and Mbd3 depletion rescued neurogenesis defects seen in Smek1/2 knockout mice. These results reveal a novel molecular mechanism underlying Smek/Mbd3/NuRD axis-mediated control of NPCs' self-renewal and neuronal differentiation during mammalian corticogenesis.
Collapse
Affiliation(s)
- Byoung-San Moon
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Hyung-Mun Yun
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Wen-Hsuan Chang
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Bradford H. Steele
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Mingyang Cai
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Si Ho Choi
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Wange Lu
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
14
|
Di Re J, Wadsworth PA, Laezza F. Intracellular Fibroblast Growth Factor 14: Emerging Risk Factor for Brain Disorders. Front Cell Neurosci 2017; 11:103. [PMID: 28469558 PMCID: PMC5396478 DOI: 10.3389/fncel.2017.00103] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/28/2017] [Indexed: 01/31/2023] Open
Abstract
The finely tuned regulation of neuronal firing relies on the integrity of ion channel macromolecular complexes. Minimal disturbances of these tightly regulated networks can lead to persistent maladaptive plasticity of brain circuitry. The intracellular fibroblast growth factor 14 (FGF14) belongs to the nexus of proteins interacting with voltage-gated Na+ (Nav) channels at the axonal initial segment. Through isoform-specific interactions with the intracellular C-terminal tail of neuronal Nav channels (Nav1.1, Nav1.2, Nav1.6), FGF14 controls channel gating, axonal targeting and phosphorylation in neurons effecting excitability. FGF14 has been also involved in synaptic transmission, plasticity and neurogenesis in the cortico-mesolimbic circuit with cognitive and affective behavioral outcomes. In translational studies, interest in FGF14 continues to rise with a growing list of associative links to diseases of the cognitive and affective domains such as neurodegeneration, depression, anxiety, addictive behaviors and recently schizophrenia, suggesting its role as a converging node in the etiology of complex brain disorders. Yet, a full understanding of FGF14 function in neurons is far from being complete and likely to involve other functions unrelated to the direct regulation of Nav channels. The goal of this Mini Review article is to provide a summary of studies on the emerging role of FGF14 in complex brain disorders.
Collapse
Affiliation(s)
- Jessica Di Re
- Neuroscience Graduate Program, University of Texas Medical BranchGalveston, TX, USA.,Department of Pharmacology and Toxicology, University of Texas Medical BranchGalveston, TX, USA
| | - Paul A Wadsworth
- Biochemistry and Molecular Biology Graduate Program, The University of Texas Medical BranchGalveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical BranchGalveston, TX, USA.,Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical BranchGalveston, TX, USA.,Center for Addiction Research, The University of Texas Medical BranchGalveston, TX, USA
| |
Collapse
|
15
|
Vadodaria KC, Yanpallewar SU, Vadhvani M, Toshniwal D, Liles LC, Rommelfanger KS, Weinshenker D, Vaidya VA. Noradrenergic regulation of plasticity marker expression in the adult rodent piriform cortex. Neurosci Lett 2017; 644:76-82. [PMID: 28237805 DOI: 10.1016/j.neulet.2017.02.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 01/20/2023]
Abstract
The adult rodent piriform cortex has been reported to harbor immature neurons that express markers associated with neurodevelopment and plasticity, namely polysialylated neural cell adhesion molecule (PSA-NCAM) and doublecortin (DCX). We characterized the expression of PSA-NCAM and DCX across the rostrocaudal axis of the rat piriform cortex and observed higher numbers of PSA-NCAM and DCX positive cells in the posterior subdivision. As observed in the rat piriform cortex, Nestin-GFP reporter mice also revealed a similar gradient of GFP-positive cells with an increasing rostro-caudal gradient of expression. Given the extensive noradrenergic innervation of the piriform cortex and its role in regulating piriform cortex function and synaptic plasticity, we addressed the influence of norepinephrine (NE) on piriform cortex plasticity marker expression. Depletion of NE by treatment with the noradrenergic neurotoxin DSP-4 significantly increased the number of DCX and PSA-NCAM immunopositive cells in the piriform cortex of adult rats. Similarly, DSP-4 treated Nestin-GFP reporter mice revealed a robust induction of GFP-positive cells within the piriform cortex following NE depletion. Genetic loss of NE in dopamine β-hydroxylase knockout (Dbh -/-) mice phenocopied the effects of DSP-4, with an increase noted in PSA-NCAM and DCX positive cells in the piriform cortex. Further, chronic α2-adrenergic receptor stimulation with the agonist guanabenz increased PSA-NCAM and DCX positive cells in the piriform cortex of adult rats and GFP-positive cells in the piriform cortex of Nestin-GFP mice. By contrast, chronic α2-adrenergic receptor blockade with the antagonist yohimbine reduced PSA-NCAM and DCX positive cells in the piriform cortex of adult rats. Our results provide novel evidence for a role of NE in regulating the expression of plasticity markers, including PSA-NCAM, DCX, and nestin, within the adult mouse and rat piriform cortex.
Collapse
Affiliation(s)
- Krishna C Vadodaria
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India, India
| | - Sudhirkumar U Yanpallewar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India, India
| | - Mayur Vadhvani
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India, India
| | - Devyani Toshniwal
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India, India
| | - L Cameron Liles
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA, USA
| | - Karen S Rommelfanger
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA, USA
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India, India.
| |
Collapse
|
16
|
Alshammari MA, Alshammari TK, Nenov MN, Scala F, Laezza F. Fibroblast Growth Factor 14 Modulates the Neurogenesis of Granule Neurons in the Adult Dentate Gyrus. Mol Neurobiol 2015; 53:7254-7270. [PMID: 26687232 DOI: 10.1007/s12035-015-9568-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/29/2015] [Indexed: 11/25/2022]
Abstract
Adult neurogenesis, the production of mature neurons from progenitor cells in the adult mammalian brain, is linked to the etiology of neurodegenerative and psychiatric disorders. However, a thorough understanding of the molecular elements at the base of adult neurogenesis remains elusive. Here, we provide evidence for a previously undescribed function of fibroblast growth factor 14 (FGF14), a brain disease-associated factor that controls neuronal excitability and synaptic plasticity, in regulating adult neurogenesis in the dentate gyrus (DG). We found that FGF14 is dynamically expressed in restricted subtypes of sex determining region Y-box 2 (Sox2)-positive and doublecortin (DCX)-positive neural progenitors in the DG. Bromodeoxyuridine (BrdU) incorporation studies and confocal imaging revealed that genetic deletion of Fgf14 in Fgf14 -/- mice leads to a significant change in the proportion of proliferating and immature and mature newly born adult granule cells. This results in an increase in the late immature and early mature population of DCX and calretinin (CR)-positive neurons. Electrophysiological extracellular field recordings showed reduced minimal threshold response and impaired paired-pulse facilitation at the perforant path to DG inputs in Fgf14 -/- compared to Fgf14 +/+ mice, supporting disrupted synaptic connectivity as a correlative read-out to impaired neurogenesis. These new insights into the biology of FGF14 in neurogenesis shed light into the signaling pathways associated with disrupted functions in complex brain diseases.
Collapse
Affiliation(s)
- Musaad A Alshammari
- Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX, USA
- Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Tahani K Alshammari
- Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX, USA
- Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Miroslav N Nenov
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Federico Scala
- Biophysics Graduate Program, Institute of Human Physiology, Università Cattolica, Rome, Italy
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Fernanda Laezza
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA.
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA.
- Center for Biomedical Engineering, The University of Texas Medical Branch, Galveston, TX, USA.
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Medical Research Building 7.102B, 301 University Boulevard, Galveston, TX, 77555, USA.
| |
Collapse
|
17
|
Suarez-Mier GB, Buckwalter MS. Glial Fibrillary Acidic Protein-Expressing Glia in the Mouse Lung. ASN Neuro 2015; 7:7/5/1759091415601636. [PMID: 26442852 PMCID: PMC4601129 DOI: 10.1177/1759091415601636] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Autonomic nerves regulate important functions in visceral organs, including the lung. The postganglionic portion of these nerves is ensheathed by glial cells known as non-myelinating Schwann cells. In the brain, glia play important functional roles in neurotransmission, neuroinflammation, and maintenance of the blood brain barrier. Similarly, enteric glia are now known to have analogous roles in gastrointestinal neurotransmission, inflammatory response, and barrier formation. In contrast to this, very little is known about the function of glia in other visceral organs. Like the gut, the lung forms a barrier between airborne pathogens and the bloodstream, and autonomic lung innervation is known to affect pulmonary inflammation and lung function. Lung glia are described as non-myelinating Schwann cells but their function is not known, and indeed no transgenic tools have been validated to study them in vivo. The primary goal of this research was, therefore, to investigate the relationship between non-myelinating Schwann cells and pulmonary nerves in the airways and vasculature and to validate existing transgenic mouse tools that would be useful for studying their function. We focused on the glial fibrillary acidic protein promoter, which is a cognate marker of astrocytes that is expressed by enteric glia and non-myelinating Schwann cells. We describe the morphology of non-myelinating Schwann cells in the lung and verify that they express glial fibrillary acidic protein and S100, a classic glial marker. Furthermore, we characterize the relationship of non-myelinating Schwann cells to pulmonary nerves. Finally, we report tools for studying their function, including a commercially available transgenic mouse line.
Collapse
Affiliation(s)
- Gabriela B Suarez-Mier
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, USA Stanford Neurosciences Institute, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, USA Department of Neurosurgery, Stanford Medical School, Stanford, CA, USA
| |
Collapse
|
18
|
Contet C, Kim A, Le D, Iyengar SK, Kotzebue RW, Yuan CJ, Kieffer BL, Mandyam CD. μ-Opioid receptors mediate the effects of chronic ethanol binge drinking on the hippocampal neurogenic niche. Addict Biol 2014; 19:770-80. [PMID: 23461397 DOI: 10.1111/adb.12040] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Ethanol exposure and withdrawal alter the generation of new neurons in the adult hippocampus. The endogenous opioid system, particularly the μ-opioid receptor (MOR), can modulate neural progenitors and also plays a critical role in ethanol drinking and dependence. In the present study, we sought to determine whether MOR contributes to the effects of ethanol on the dentate gyrus (DG) neurogenic niche. MOR wild-type (WT), heterozygous (Het) and knockout (KO) littermates were subjected to voluntary ethanol drinking in repeated limited-access two-bottle choice (2BC) sessions. MOR deficiency did not alter progenitor proliferation, neuronal differentiation and maturation, apoptosis or microglia in ethanol-naïve mice. When exposed to five consecutive weeks of 2BC, MOR mutant mice exhibited a gene-dosage-dependent reduction of ethanol consumption compared with WT mice. Introducing a week of ethanol deprivation between each week of 2BC increased ethanol consumption in all genotypes and produced equivalent intakes in WT, Het and KO mice. Under the latter paradigm, ethanol drinking decreased progenitor proliferation and neuronal differentiation in the DG of WT mice. Interestingly, WT mice exhibited a strong negative correlation between ethanol intake and proliferation, which was disrupted in Het and KO mice. Moreover, MOR deficiency blocked the effect of ethanol on neuronal differentiation. MOR deficiency also protected against the neuroimmune response to ethanol drinking. Finally, chronic binge drinking induced a paradoxical decrease in apoptosis, which was independent of MOR. Altogether, our data suggest that MOR is implicated in some of the neuroplastic changes produced by chronic ethanol exposure in the DG.
Collapse
Affiliation(s)
- Candice Contet
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | - Airee Kim
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | - David Le
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | - Siddharth K. Iyengar
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | - Roxanne W. Kotzebue
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | - Clara J. Yuan
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla CA USA
| | - Brigitte L. Kieffer
- Département Neurobiologie; Institut de Génétique et de Biologie Moléculaire et Cellulaire; Centre National de Recherche Scientifique/Institut National de la Santé et de la Recherche Médicale/Université de Strasbourg; France
| | - Chitra D. Mandyam
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California San Diego; La Jolla CA USA
| |
Collapse
|
19
|
Poon A, Goldowitz D. Identification of genetic loci that modulate cell proliferation in the adult rostral migratory stream using the expanded panel of BXD mice. BMC Genomics 2014; 15:206. [PMID: 24640950 PMCID: PMC4004255 DOI: 10.1186/1471-2164-15-206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 03/10/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Adult neurogenesis, which is the continual production of new neurons in the mature brain, demonstrates the strikingly plastic nature of the nervous system. Adult neural stem cells and their neural precursors, collectively referred to as neural progenitor cells (NPCs), are present in the subgranular zone (SGZ) of the dentate gyrus, the subventricular zone (SVZ), and rostral migratory stream (RMS). In order to harness the potential of NPCs to treat neurodegenerative diseases and brain injuries, it will be important to understand the molecules that regulate NPCs in the adult brain. The genetic basis underlying NPC proliferation is still not fully understood. From our previous quantitative trait locus (QTL) analysis, we had success in using a relatively small reference population of recombinant inbred strains of mice (AXBXA) to identify a genetic region that is significantly correlated with NPC proliferation in the RMS. RESULTS In this study, we expanded our initial QTL mapping of RMS proliferation to a far richer genetic resource, the BXD RI mouse strains. A 3-fold difference in the number of proliferative, bromodeoxyuridine (BrdU)-labeled cells was quantified in the adult RMS of 61 BXD RI strains. RMS cell proliferation is highly dependent on the genetic background of the mice with an estimated heritability of 0.58. Genome-wide mapping revealed a significant QTL on chromosome (Chr) 6 and a suggestive QTL on Chr 11 regulating the number of NPCs in the RMS. Composite interval analysis further revealed secondary QTLs on Chr 14 and Chr 18. The loci regulating RMS cell proliferation did not overlap with the suggestive loci modulating cell proliferation in the SGZ. These mapped loci serve as starting points to identify genes important for this process. A subset of candidate genes in this region is associated with cell proliferation and neurogenesis. Interconnectivity of these candidate genes was demonstrated using pathway and transcriptional covariance analyses. CONCLUSIONS Differences in RMS cell proliferation across the BXD RI strains identifies genetic loci that serve to provide insights into the interplay of underlying genes that may be important for regulating NPC proliferation in the adult mouse brain.
Collapse
Affiliation(s)
| | - Daniel Goldowitz
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.
| |
Collapse
|
20
|
Hattori T, Shimizu S, Koyama Y, Emoto H, Matsumoto Y, Kumamoto N, Yamada K, Takamura H, Matsuzaki S, Katayama T, Tohyama M, Ito A. DISC1 (disrupted-in-schizophrenia-1) regulates differentiation of oligodendrocytes. PLoS One 2014; 9:e88506. [PMID: 24516667 PMCID: PMC3917910 DOI: 10.1371/journal.pone.0088506] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 01/08/2014] [Indexed: 02/05/2023] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a gene disrupted by a translocation, t(1;11) (q42.1;q14.3), that segregates with major psychiatric disorders, including schizophrenia, recurrent major depression and bipolar affective disorder, in a Scottish family. Here we report that mammalian DISC1 endogenously expressed in oligodendroglial lineage cells negatively regulates differentiation of oligodendrocyte precursor cells into oligodendrocytes. DISC1 expression was detected in oligodendrocytes of the mouse corpus callosum at P14 and P70. DISC1 mRNA was expressed in primary cultured rat cortical oligodendrocyte precursor cells and decreased when oligodendrocyte precursor cells were induced to differentiate by PDGF deprivation. Immunocytochemical analysis showed that overexpressed DISC1 was localized in the cell bodies and processes of oligodendrocyte precursor cells and oligodendrocytes. We show that expression of the myelin related markers, CNPase and MBP, as well as the number of cells with a matured oligodendrocyte morphology, were decreased following full length DISC1 overexpression. Conversely, both expression of CNPase and the number of oligodendrocytes with a mature morphology were increased following knockdown of endogenous DISC1 by RNA interference. Overexpression of a truncated form of DISC1 also resulted in an increase in expression of myelin related proteins and the number of mature oligodendrocytes, potentially acting via a dominant negative mechanism. We also identified involvement of Sox10 and Nkx2.2 in the DISC1 regulatory pathway of oligodendrocyte differentiation, both well-known transcription factors involved in the regulation of myelin genes.
Collapse
Affiliation(s)
- Tsuyoshi Hattori
- Department of Molecular Neuropsychiatry, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- * E-mail:
| | - Shoko Shimizu
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Sayama, Osaka, Japan
| | - Yoshihisa Koyama
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hisayo Emoto
- Pharmacology Research Laboratories, Dainippon Sumitomo Pharma Co, Ltd, Suita, Osaka, Japan
| | - Yuji Matsumoto
- Pharmacology Research Laboratories, Dainippon Sumitomo Pharma Co, Ltd, Suita, Osaka, Japan
| | - Natsuko Kumamoto
- Department of Neurobiology and Anatomy, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Kohei Yamada
- Department of Child Development & Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of Medicine, Suita, Osaka, Japan
| | - Hironori Takamura
- Department of Child Development & Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of Medicine, Suita, Osaka, Japan
| | - Shinsuke Matsuzaki
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Child Development & Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of Medicine, Suita, Osaka, Japan
| | - Taiichi Katayama
- Department of Child Development & Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of Medicine, Suita, Osaka, Japan
| | - Masaya Tohyama
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Child Development & Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University and Hamamatsu University School of Medicine, Suita, Osaka, Japan
- Division of Molecular Brain Science, Research Institute of Traditional Asian Medicine, Kinki University, Sayama, Osaka, Japan
| | - Akira Ito
- Department of Molecular Neuropsychiatry, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
21
|
Bonini SA, Ferrari-Toninelli G, Montinaro M, Memo M. Notch signalling in adult neurons: a potential target for microtubule stabilization. Ther Adv Neurol Disord 2013; 6:375-85. [PMID: 24228073 DOI: 10.1177/1756285613490051] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cytoskeletal dysfunction has been proposed during the last decade as one of the main mechanisms involved in the aetiology of several neurodegenerative diseases. Microtubules are basic elements of the cytoskeleton and the dysregulation of microtubule stability has been demonstrated to be causative for axonal transport impairment, synaptic contact degeneration, impaired neuronal function leading finally to neuronal loss. Several pathways are implicated in the microtubule assembly/disassembly process. Emerging evidence is focusing on Notch as a microtubule dynamics regulator. We demonstrated that activation of Notch signalling results in increased microtubule stability and changes in axonal morphology and branching. By contrast, Notch inhibition leads to an increase in cytoskeleton plasticity with intense neurite remodelling. Until now, several microtubule-binding compounds have been tested and the results have provided proof of concept that microtubule-binding agents or compounds with the ability to stabilize microtubules may have therapeutic potential for the treatment of Alzheimer's disease and other neurodegenerative diseases. In this review, based on its key role in cytoskeletal dynamics modulation, we propose Notch as a new potential target for microtubule stabilization.
Collapse
Affiliation(s)
- Sara Anna Bonini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | | | |
Collapse
|
22
|
Effects of Treadmill Exercise on Neural Stem Cells, Cell Proliferation, and Neuroblast Differentiation in the Subgranular Zone of the Dentate Gyrus in Cyclooxygenase-2 Knockout Mice. Neurochem Res 2013; 38:2559-69. [DOI: 10.1007/s11064-013-1169-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/25/2013] [Accepted: 09/27/2013] [Indexed: 01/26/2023]
|
23
|
Zhuang PW, Cui GZ, Zhang YJ, Zhang MX, Guo H, Zhang JB, Lu ZQ, Isaiah AO, Lin YX. Baicalin regulates neuronal fate decision in neural stem/progenitor cells and stimulates hippocampal neurogenesis in adult rats. CNS Neurosci Ther 2013; 19:154-62. [PMID: 23302221 DOI: 10.1111/cns.12050] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/19/2012] [Accepted: 11/20/2012] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Recent studies revealed that baicalin, a flavonoid compound derived from the root of Scutellaria baicalensis Georgi, could promote neuron differentiation of NSPCs after commencing the differentiation process in vitro. However, this may not be the most efficacious strategy to determinate cell fate. Here, we have investigated whether baicalin can influence early events of neuron generation and stimulate adult neurogenesis. RESULTS Transient exposure of NSPCs to baicalin during proliferation could activate Mash1 to alter the differential fate and increase the proportion of cells expressing neuronal markers. Seven days after, rats were exposed to transient cerebral ischemia, they were treated for 3 weeks with baicalin, BrdU labeling study showed that exposure to baicalin increased the number of newly generated cells in hippocampus, BrdU/NeuN double staining analysis indicated that baicalin could promote new neuron production after cerebral ischemia. Additionally, Morris water maze test showed that delayed postischemic treatment with baicalin improved cognitive impairment. CONCLUSIONS These results identify the existence of a single molecule, baicalin, which can specify the neuronal fate of multipotent NSPCs and stimulate neurogenesis, making it a promising candidate for developing clinically relevant strategies to manipulate neuronal fate of NSPCs for brain repair.
Collapse
Affiliation(s)
- Peng-Wei Zhuang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Desplats P, Spencer B, Crews L, Pathel P, Morvinski-Friedmann D, Kosberg K, Roberts S, Patrick C, Winner B, Winkler J, Masliah E. α-Synuclein induces alterations in adult neurogenesis in Parkinson disease models via p53-mediated repression of Notch1. J Biol Chem 2012; 287:31691-702. [PMID: 22833673 DOI: 10.1074/jbc.m112.354522] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease is characterized by the loss of dopaminergic neurons mainly in the substantia nigra. Accumulation of α-synuclein and cell loss has been also reported in many other brain regions including the hippocampus, where it might impair adult neurogenesis, contributing to nonmotor symptoms. However, the molecular mechanisms of these alterations are still unknown. In this report we show that α-synuclein-accumulating adult rat hippocampus neural progenitors present aberrant neuronal differentiation, with reduction of Notch1 expression and downstream signaling targets. We characterized a Notch1 proximal promoter that contains p53 canonical response elements. In vivo binding of p53 represses the transcription of Notch1 in neurons. Moreover, we demonstrated that α-synuclein directly binds to the DNA at Notch1 promoter vicinity and also interacts with p53 protein, facilitating or increasing Notch1 signaling repression, which interferes with maturation and survival of neural progenitors cells. This study provides a molecular basis for α-synuclein-mediated disruption of adult neurogenesis in Parkinson disease.
Collapse
Affiliation(s)
- Paula Desplats
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California 92093, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhuang P, Zhang Y, Cui G, Bian Y, Zhang M, Zhang J, Liu Y, Yang X, Isaiah AO, Lin Y, Jiang Y. Direct stimulation of adult neural stem/progenitor cells in vitro and neurogenesis in vivo by salvianolic acid B. PLoS One 2012; 7:e35636. [PMID: 22545124 PMCID: PMC3335811 DOI: 10.1371/journal.pone.0035636] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/19/2012] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Small molecules have been shown to modulate the neurogenesis processes. In search for new therapeutic drugs, the herbs used in traditional medicines for neurogenesis are promising candidates. METHODOLOGY AND PRINCIPAL FINDINGS We selected a total of 45 natural compounds from Traditional Chinese herbal medicines which are extensively used in China to treat stroke clinically, and tested their proliferation-inducing activities on neural stem/progenitor cells (NSPCs). The screening results showed that salvianolic acid B (Sal B) displayed marked effects on the induction of proliferation of NSPCs. We further demonstrated that Sal B promoted NSPCs proliferation in dose- and time-dependent manners. To explore the molecular mechanism, PI3K/Akt, MEK/ERK and Notch signaling pathways were investigated. Cell proliferation assay demonstrated that Ly294002 (PI3K/Akt inhibitor), but neither U0126 (ERK inhibitor) nor DAPT (Notch inhibitor) inhibited the Sal B-induced proliferation of cells. Western Blotting results showed that stimulation of NSPCs with Sal B enhanced the phosphorylation of Akt, and Ly294002 abolished this effect, confirming the role of Akt in Sal B mediated proliferation of NSPCs. Rats exposed to transient cerebral ischemia were treated for 4 weeks with Sal B from the 7th day after stroke. BrdU incorporation assay results showed that exposure Sal B could maintain the proliferation of NSPCs after cerebral ischemia. Morris water maze test showed that delayed post-ischemic treatment with Sal B improved cognitive impairment after stroke in rats. SIGNIFICANCE Sal B could maintain the NSPCs self-renew and promote proliferation, which was mediated by PI3K/Akt signal pathway. And delayed post-ischemic treatment with Sal B improved cognitive impairment after stroke in rats. These findings suggested that Sal B may act as a potential drug in treatment of brain injury or neurodegenerative diseases.
Collapse
Affiliation(s)
- Pengwei Zhuang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanjun Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- * E-mail:
| | - Guangzhi Cui
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Bian
- Chinese Medical College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mixia Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinbao Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Liu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinpeng Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Adejobi Oluwaniyi Isaiah
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingxue Lin
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yongbo Jiang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Traditional Chinese Medicine Pharmacology, Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
26
|
Sato C, Zhao G, Ilagan MXG. An overview of notch signaling in adult tissue renewal and maintenance. Curr Alzheimer Res 2012; 9:227-40. [PMID: 21605032 PMCID: PMC4361071 DOI: 10.2174/156720512799361600] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 05/27/2011] [Accepted: 06/10/2011] [Indexed: 11/22/2022]
Abstract
The Notch pathway is a critical mediator of short-range cell-cell communication that is reiteratively used to regulate a diverse array of cellular processes during embryonic development and the renewal and maintenance of adult tissues. Most Notch-dependent processes utilize a core signaling mechanism that is dependent on regulated intramembrane proteolysis: Upon ligand binding, Notch receptors undergo ectodomain shedding by ADAM metalloproteases, followed by γ-secretase-mediated intramembrane proteolysis. This releases the Notch intracellular domain, which translocates to the nucleus to activate transcription. In this review, we highlight the roles of Notch signaling particularly in self-renewing tissues in adults and several human diseases and raise some key considerations when targeting ADAMs and γ-secretase as disease-modifying strategies for Alzheimer's Disease.
Collapse
Affiliation(s)
- Chihiro Sato
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA 63110
| | - Guojun Zhao
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA 63110
| | - Ma. Xenia G. Ilagan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA 63110
| |
Collapse
|
27
|
Mandyam CD, Koob GF. The addicted brain craves new neurons: putative role for adult-born progenitors in promoting recovery. Trends Neurosci 2012; 35:250-60. [PMID: 22265158 DOI: 10.1016/j.tins.2011.12.005] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 08/30/2011] [Accepted: 12/13/2011] [Indexed: 02/08/2023]
Abstract
Addiction is a chronic relapsing disorder associated with compulsive drug taking, drug seeking and a loss of control in limiting intake, reflected in three stages of a recurrent cycle: binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation ("craving"). This review discusses the role of adult-born neural and glial progenitors in drug seeking associated with the different stages of the addiction cycle. A review of the current literature suggests that the loss of newly born progenitors, particularly in hippocampal and cortical regions, plays a role in determining vulnerability to relapse in rodent models of drug addiction. The normalization of drug-impaired neurogenesis or gliogenesis may help reverse neuroplasticity during abstinence and, thus, may help reduce the vulnerability to relapse and aid recovery.
Collapse
Affiliation(s)
- Chitra D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA.
| | | |
Collapse
|
28
|
Masiulis I, Yun S, Eisch AJ. The interesting interplay between interneurons and adult hippocampal neurogenesis. Mol Neurobiol 2011; 44:287-302. [PMID: 21956642 DOI: 10.1007/s12035-011-8207-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 09/07/2011] [Indexed: 10/17/2022]
Abstract
Adult neurogenesis is a unique form of plasticity found in the hippocampus, a brain region key to learning and memory formation. While many external stimuli are known to modulate the generation of new neurons in the hippocampus, little is known about the local circuitry mechanisms that regulate the process of adult neurogenesis. The neurogenic niche in the hippocampus is highly complex and consists of a heterogeneous population of cells including interneurons. Because interneurons are already highly integrated into the hippocampal circuitry, they are in a prime position to influence the proliferation, survival, and maturation of adult-generated cells in the dentate gyrus. Here, we review the current state of our understanding on the interplay between interneurons and adult hippocampal neurogenesis. We focus on activity- and signaling-dependent mechanisms, as well as research on human diseases that could provide better insight into how interneurons in general might add to our comprehension of the regulation and function of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Irene Masiulis
- UT Southwestern Medical Center, Dallas, TX 75390-9070, USA.
| | | | | |
Collapse
|
29
|
Ables JL, Breunig JJ, Eisch AJ, Rakic P. Not(ch) just development: Notch signalling in the adult brain. Nat Rev Neurosci 2011; 12:269-83. [PMID: 21505516 DOI: 10.1038/nrn3024] [Citation(s) in RCA: 325] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The Notch pathway is often regarded as a developmental pathway, but components of Notch signalling are expressed and active in the adult brain. With the advent of more sophisticated genetic manipulations, evidence has emerged that suggests both conserved and novel roles for Notch signalling in the adult brain. Not surprisingly, Notch is a key regulator of adult neural stem cells, but it is increasingly clear that Notch signalling also has roles in the regulation of migration, morphology, synaptic plasticity and survival of immature and mature neurons. Understanding the many functions of Notch signalling in the adult brain, and its dysfunction in neurodegenerative disease and malignancy, is crucial to the development of new therapeutics that are centred around this pathway.
Collapse
Affiliation(s)
- Jessica L Ables
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
30
|
Roitbak T, Thomas K, Martin A, Allan A, Cunningham LA. Moderate fetal alcohol exposure impairs neurogenic capacity of murine neural stem cells isolated from the adult subventricular zone. Exp Neurol 2011; 229:522-5. [PMID: 21419122 DOI: 10.1016/j.expneurol.2011.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 02/11/2011] [Accepted: 03/04/2011] [Indexed: 12/31/2022]
Abstract
Gestational alcohol exposure leads to a spectrum of neurological symptoms which range from severe mental retardation caused by high dose exposure, to subtle cognitive and neuropsychiatric symptoms caused by low-to-moderate doses. We and other investigators have demonstrated that exposure to moderate levels of alcohol throughout gestation leads to impaired neurogenesis in the adult hippocampus, although the mechanisms by which this occurs are not known. To begin to distinguish cell-intrinsic from microenvironmental contributions to impaired adult neurogenesis, we isolated neural stem progenitor cells (NSPCs) from the adult SVZ of mice exposed to moderate levels of alcohol throughout gestation. We found that NSPCs isolated from fetal alcohol exposed (FAE) mice displayed reduced neurosphere formation in culture, as assessed by a serial passage neurosphere assay, and reduced neuronal differentiation upon growth factor withdrawal. These studies suggest that gestational alcohol exposure leads to long-lasting NSPC-intrinsic dysregulation, which may underlie in vivo neurogenic deficits.
Collapse
Affiliation(s)
- Tamara Roitbak
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131–0001, USA
| | | | | | | | | |
Collapse
|
31
|
Merz K, Herold S, Lie DC. CREB in adult neurogenesis - master and partner in the development of adult-born neurons? Eur J Neurosci 2011; 33:1078-86. [DOI: 10.1111/j.1460-9568.2011.07606.x] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
32
|
|
33
|
Kim YH, Chung JI, Woo HG, Jung YS, Lee SH, Moon CH, Suh-Kim H, Baik EJ. Differential regulation of proliferation and differentiation in neural precursor cells by the Jak pathway. Stem Cells 2011; 28:1816-28. [PMID: 20979137 DOI: 10.1002/stem.511] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neuronal precursor cells (NPCs) are temporally regulated and have the ability to proliferate and differentiate into mature neurons, oligodendrocytes, and astrocytes in the presence of growth factors (GFs). In the present study, the role of the Jak pathway in brain development was investigated in NPCs derived from neurosphere cultures using Jak2 and Jak3 small interfering RNAs and specific inhibitors. Jak2 inhibition profoundly decreased NPC proliferation, preventing further differentiation into neurons and glial cells. However, Jak3 inhibition induced neuronal differentiation accompanied by neurite growth. This phenomenon was due to the Jak3 inhibition-mediated induction of neurogenin (Ngn)2 and NeuroD in NPCs. Jak3 inhibition induced NPCs to differentiate into scattered neurons and increased the expression of Tuj1, microtubule associated protein 2 (MAP2), Olig2, and neuroglial protein (NG)2, but decreased glial fibrillary acidic protein (GFAP) expression, with predominant neurogenesis/polydendrogenesis compared with astrogliogenesis. Therefore, Jak2 may be important for NPC proliferation and maintenance, whereas knocking-down of Jak3 signaling is essential for NPC differentiation into neurons and oligodendrocytes but does not lead to astrocyte differentiation. These results suggest that NPC proliferation and differentiation are differentially regulated by the Jak pathway.
Collapse
Affiliation(s)
- Yun Hee Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Varela-Nallar L, Aranguiz FC, Abbott AC, Slater PG, Inestrosa NC. Adult hippocampal neurogenesis in aging and Alzheimer's disease. ACTA ACUST UNITED AC 2010; 90:284-96. [DOI: 10.1002/bdrc.20193] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
Lee MH, Wang T, Jang MH, Steiner J, Haughey N, Ming GL, Song H, Nath A, Venkatesan A. Rescue of adult hippocampal neurogenesis in a mouse model of HIV neurologic disease. Neurobiol Dis 2010; 41:678-87. [PMID: 21146610 DOI: 10.1016/j.nbd.2010.12.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 11/16/2010] [Accepted: 12/02/2010] [Indexed: 02/02/2023] Open
Abstract
The prevalence of central nervous system (CNS) neurologic dysfunction associated with human immunodeficiency virus (HIV) infection continues to increase, despite the use of antiretroviral therapy. Previous work has focused on the deleterious effects of HIV on mature neurons and on development of neuroprotective strategies, which have consistently failed to show a meaningful clinical benefit. It is now well established that new neurons are continuously generated in discrete regions in the adult mammalian brain, and accumulating evidence supports important roles for these neurons in specific cognitive functions. In a transgenic mouse model of HIV neurologic disease with glial expression of the HIV envelope protein gp120, we demonstrate a significant reduction in proliferation of hippocampal neural progenitors in the dentate gyrus of adult animals, resulting in a dramatic decrease in the number of newborn neurons in the adult brain. We identify amplifying neural progenitor cells (ANPs) as the first class of progenitors affected by gp120, and we also demonstrate that newly generated neurons exhibit aberrant dendritic development. Furthermore, voluntary exercise and treatment with a selective serotonin reuptake inhibitor increase the ANP population and rescue the observed deficits in gp120 transgenic mice. Thus, during HIV infection, the envelope protein gp120 may potently inhibit adult hippocampal neurogenesis, and neurorestorative approaches may be effective in ameliorating these effects. Our study has significant implications for the development of novel therapeutic approaches for HIV-infected individuals with neurologic dysfunction and may be applicable to other neurodegenerative diseases in which hippocampal neurogenesis is impaired.
Collapse
Affiliation(s)
- Myoung-Hwa Lee
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Meyer 6-113, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Poon A, Li Z, Wolfe GW, Lu L, Williams RW, Hayes NL, Nowakowski RS, Goldowitz D. Identification of a Chr 11 quantitative trait locus that modulates proliferation in the rostral migratory stream of the adult mouse brain. Eur J Neurosci 2010; 32:523-37. [PMID: 20718853 DOI: 10.1111/j.1460-9568.2010.07316.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neuron production takes place continuously in the rostral migratory stream (RMS) of the adult mammalian brain. The molecular mechanisms that regulate progenitor cell division and differentiation in the RMS remain largely unknown. Here, we surveyed the mouse genome in an unbiased manner to identify candidate gene loci that regulate proliferation in the adult RMS. We quantified neurogenesis in adult C57BL/6J and A/J mice, and 27 recombinant inbred lines derived from those parental strains. We showed that the A/J RMS had greater numbers of bromodeoxyuridine-labeled cells than that of C57BL/6J mice with similar cell cycle parameters, indicating that the differences in the number of bromodeoxyuridine-positive cells reflected the number of proliferating cells between the strains. AXB and BXA recombinant inbred strains demonstrated even greater variation in the numbers of proliferating cells. Genome-wide mapping of this trait revealed that chromosome 11 harbors a significant quantitative trait locus at 116.75 +/- 0.75 Mb that affects cell proliferation in the adult RMS. The genomic regions that influence RMS proliferation did not overlap with genomic regions regulating proliferation in the adult subgranular zone of the hippocampal dentate gyrus. On the contrary, a different, suggestive locus that modulates cell proliferation in the subgranular zone was mapped to chromosome 3 at 102 +/- 7 Mb. A subset of genes in the chromosome 11 quantitative trait locus region is associated with neurogenesis and cell proliferation. Our findings provide new insights into the genetic control of neural proliferation and an excellent starting point to identify genes critical to this process.
Collapse
Affiliation(s)
- Anna Poon
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Ohira K, Hagihara H, Toyama K, Takao K, Kanai M, Funakoshi H, Nakamura T, Miyakawa T. Expression of tryptophan 2,3-dioxygenase in mature granule cells of the adult mouse dentate gyrus. Mol Brain 2010; 3:26. [PMID: 20815922 PMCID: PMC2945337 DOI: 10.1186/1756-6606-3-26] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 09/05/2010] [Indexed: 12/20/2022] Open
Abstract
New granule cells are continuously generated in the dentate gyrus of the adult hippocampus. During granule cell maturation, the mechanisms that differentiate new cells not only describe the degree of cell differentiation, but also crucially regulate the progression of cell differentiation. Here, we describe a gene, tryptophan 2,3-dioxygenase (TDO), whose expression distinguishes stem cells from more differentiated cells among the granule cells of the adult mouse dentate gyrus. The use of markers for proliferation, neural progenitors, and immature and mature granule cells indicated that TDO was expressed in mature cells and in some immature cells. In mice heterozygous for the alpha-isoform of calcium/calmodulin-dependent protein kinase II, in which dentate gyrus granule cells fail to mature normally, TDO immunoreactivity was substantially downregulated in the dentate gyrus granule cells. Moreover, a 5-bromo-2'-deoxyuridine labeling experiment revealed that new neurons began to express TDO between 2 and 4 wk after the neurons were generated, when the axons and dendrites of the granule cells developed and synaptogenesis occurred. These findings indicate that TDO might be required at a late-stage of granule cell development, such as during axonal and dendritic growth, synaptogenesis and its maturation.
Collapse
Affiliation(s)
- Koji Ohira
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Stevens HE, Smith KM, Rash BG, Vaccarino FM. Neural stem cell regulation, fibroblast growth factors, and the developmental origins of neuropsychiatric disorders. Front Neurosci 2010; 4. [PMID: 20877431 PMCID: PMC2944667 DOI: 10.3389/fnins.2010.00059] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2010] [Accepted: 07/20/2010] [Indexed: 12/15/2022] Open
Abstract
There is increasing appreciation for the neurodevelopmental underpinnings of many psychiatric disorders. Disorders that begin in childhood such as autism, language disorders or mental retardation as well as adult-onset mental disorders may have origins early in neurodevelopment. Neural stem cells (NSCs) can be defined as self-renewing, multipotent cells that are present in both the embryonic and adult brain. Several recent research findings demonstrate that psychiatric illness may begin with abnormal specification, growth, expansion and differentiation of embryonic NSCs. For example, candidate susceptibility genes for schizophrenia, autism and major depression include the signaling molecule Disrupted In Schizophrenia-1 (DISC-1), the homeodomain gene engrailed-2 (EN-2), and several receptor tyrosine kinases, including brain-derived growth factor and fibroblast growth factors, all of which have been shown to play important roles in NSCs or neuronal precursors. We will discuss here stem cell biology, signaling factors that affect these cells, and the potential contribution of these processes to the etiology of neuropsychiatric disorders. Hypotheses about how some of these factors relate to psychiatric disorders will be reviewed.
Collapse
Affiliation(s)
- Hanna E Stevens
- Yale Child Study Center, Yale University School of Medicine New Haven, CT, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Notch1 regulates neural stem cell (NSC) number during development, but its role in adult neurogenesis is unclear. We generated nestin-CreER(T2)/R26R-YFP/Notch1(loxP/loxP) [Notch1inducible knock-out (iKO)] mice to allow tamoxifen (TAM)-inducible elimination of Notch1 and concomitant expression of yellow fluorescent protein (YFP) in nestin-expressing Type-1 NSCs and their progeny in the adult hippocampal subgranular zone (SGZ). Consistent with previous research, YFP+ cells in all stages of neurogenesis were evident in the subgranular zone (SGZ) of wild-type (WT) mice (nestin-CreER(T2)/R26R-YFP/Notch1(w/w)) after tamoxifen (post-TAM), producing adult-generated YFP+ dentate gyrus neurons. Compared with WT littermates, Notch1 iKO mice had similar numbers of total SGZ YFP+ cells 13 and 30 d post-TAM but had significantly fewer SGZ YFP+ cells 60 and 90 d post-TAM. Significantly fewer YFP+ Type-1 NSCs and transiently amplifying progenitors (TAPs) resulted in generation of fewer YFP+ granule neurons in Notch1 iKO mice. Strikingly, 30 d of running rescued this deficit, as the total YFP+ cell number in Notch iKO mice was equivalent to WT levels. This was even more notable given the persistent deficits in the Type-1 NSC and TAP reservoirs. Our data show that Notch1 signaling is required to maintain a reservoir of undifferentiated cells and ensure continuity of adult hippocampal neurogenesis, but that alternative Notch- and Type-1 NSC-independent pathways compensate in response to physical activity. These data shed light on the complex relationship between Type-1 NSCs, adult neurogenesis, the neurogenic niche, and environmental stimuli.
Collapse
|
40
|
Hattori T, Shimizu S, Koyama Y, Yamada K, Kuwahara R, Kumamoto N, Matsuzaki S, Ito A, Katayama T, Tohyama M. DISC1 regulates cell-cell adhesion, cell-matrix adhesion and neurite outgrowth. Mol Psychiatry 2010; 15:778, 798-809. [PMID: 20479754 DOI: 10.1038/mp.2010.60] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a promising susceptibility gene for major mental illness. Recent studies have implicated DISC1 in key neurodevelopmental processes, including neurite outgrowth, neuronal migration and proliferation. Here, we report that DISC1 regulates cell-cell and cell-matrix adhesion and neurite outgrowth. DISC1 overexpression increased expression of the adherence junction protein N-cadherin and enhanced cell-cell adhesion. The increased N-cadherin accumulated in the areas of cell-cell contact. DISC1 overexpression also enhanced cell-matrix adhesion by inducing expression of beta1-integrin protein. In the presence of nerve growth factor (NGF), DISC1 overexpression increased beta1-integrin expression at the cell membrane and growth cone. NGF-induced neurite extension was enhanced by DISC1, and anti-beta1-integrin antibody reduced the neurite outgrowth of DISC1-overexpressing cells to the control level. Furthermore, DISC1 also regulated N-cadherin and beta1-integrin expression at the cell membrane in primary neurons. We conclude that DISC1 regulates cell-cell adhesion and cell-matrix adhesion by regulating the expression of adhesion molecules.
Collapse
Affiliation(s)
- T Hattori
- Department of Molecular Neuropsychiatry, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Renault VM, Rafalski VA, Morgan AA, Salih DAM, Brett JO, Webb AE, Villeda SA, Thekkat PU, Guillerey C, Denko NC, Palmer TD, Butte AJ, Brunet A. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 2010; 5:527-39. [PMID: 19896443 DOI: 10.1016/j.stem.2009.09.014] [Citation(s) in RCA: 459] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 06/26/2009] [Accepted: 09/28/2009] [Indexed: 02/08/2023]
Abstract
In the nervous system, neural stem cells (NSCs) are necessary for the generation of new neurons and for cognitive function. Here we show that FoxO3, a member of a transcription factor family known to extend lifespan in invertebrates, regulates the NSC pool. We find that adult FoxO3(-/-) mice have fewer NSCs in vivo than wild-type counterparts. NSCs isolated from adult FoxO3(-/-) mice have decreased self-renewal and an impaired ability to generate different neural lineages. Identification of the FoxO3-dependent gene expression profile in NSCs suggests that FoxO3 regulates the NSC pool by inducing a program of genes that preserves quiescence, prevents premature differentiation, and controls oxygen metabolism. The ability of FoxO3 to prevent the premature depletion of NSCs might have important implications for counteracting brain aging in long-lived species.
Collapse
|
42
|
Nixon K, Morris SA, Liput DJ, Kelso ML. Roles of neural stem cells and adult neurogenesis in adolescent alcohol use disorders. Alcohol 2010; 44:39-56. [PMID: 20113873 DOI: 10.1016/j.alcohol.2009.11.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2009] [Revised: 10/24/2009] [Accepted: 11/07/2009] [Indexed: 01/19/2023]
Abstract
This review discusses the contributions of a newly considered form of plasticity, the ongoing production of new neurons from neural stem cells, or adult neurogenesis, within the context of neuropathologies that occur with excessive alcohol intake in the adolescents. Neural stem cells and adult neurogenesis are now thought to contribute to the structural integrity of the hippocampus, a limbic system region involved in learning, memory, behavioral control, and mood. In adolescents with alcohol use disorders (AUDs), the hippocampus appears to be particularly vulnerable to the neurodegenerative effects of alcohol, but the role of neural stem cells and adult neurogenesis in alcoholic neuropathology has only recently been considered. This review encompasses a brief overview of neural stem cells and the processes involved in adult neurogenesis, how neural stem cells are affected by alcohol, and possible differences in the neurogenic niche between adults and adolescents. Specifically, what is known about developmental differences in adult neurogenesis between the adult and adolescent is gleaned from the literature, as well as how alcohol affects this process differently among the age groups. Finally, this review suggests differences that may exist in the neurogenic niche between adults and adolescents and how these differences may contribute to the susceptibility of the adolescent hippocampus to damage. However, many more studies are needed to discern whether these developmental differences contribute to the vulnerability of the adolescent to developing an AUD.
Collapse
|
43
|
Hsieh J, Eisch AJ. Epigenetics, hippocampal neurogenesis, and neuropsychiatric disorders: unraveling the genome to understand the mind. Neurobiol Dis 2010; 39:73-84. [PMID: 20114075 DOI: 10.1016/j.nbd.2010.01.008] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 01/07/2010] [Accepted: 01/11/2010] [Indexed: 01/16/2023] Open
Abstract
In mature, differentiated neurons in the central nervous system (CNS), epigenetic mechanisms--including DNA methylation, histone modification, and regulatory noncoding RNAs--play critical roles in encoding experience and environmental stimuli into stable, behaviorally meaningful changes in gene expression. For example, epigenetic changes in mature hippocampal neurons have been implicated in learning and memory and in a variety of neuropsychiatric disorders, including depression. With all the recent (and warranted) attention given to epigenetic modifications in mature neurons, it is easy to forget that epigenetic mechanisms were initially described for their ability to promote differentiation and drive cell fate in embryonic and early postnatal development, including neurogenesis. Given the discovery of ongoing neurogenesis in the adult brain and the intriguing links among adult hippocampal neurogenesis, hippocampal function, and neuropsychiatric disorders, it is timely to complement the ongoing discussions on the role of epigenetics in mature neurons with a review on what is currently known about the role of epigenetics in adult hippocampal neurogenesis. The process of adult hippocampal neurogenesis is complex, with neural stem cells (NSCs) giving rise to fate-restricted progenitors and eventually mature dentate gyrus granule cells. Notably, neurogenesis occurs within an increasingly well-defined "neurogenic niche", where mature cellular elements like vasculature, astrocytes, and neurons release signals that can dynamically regulate neurogenesis. Here we review the evidence that key stages and aspects of adult neurogenesis are driven by epigenetic mechanisms. We discuss the intrinsic changes occurring within NSCs and their progeny that are critical for neurogenesis. We also discuss how extrinsic changes occurring in cellular components in the niche can result in altered neurogenesis. Finally we describe the potential relevance of epigenetics for understanding the relationship between hippocampal neurogenesis in neuropsychiatric disorders. We propose that a more thorough understanding of the molecular and genetic mechanisms that control the complex process of neurogenesis, including the proliferation and differentiation of NSCs, will lead to novel therapeutics for the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jenny Hsieh
- Department of Molecular Biology, University of Texas Southwestern Medical Center, TX 75390, USA.
| | | |
Collapse
|
44
|
Abstract
Stem cells are rare and unique precursor cells that participate in the building and rebuilding of tissues and organs during embryogenesis, postnatal growth, and injury repair. Stem cells are distinctively endowed with the ability to both self-renew and differentiate, such that they can replenish the stem cell pool while continuing to produce the differentiated daughter cells that are essential for tissue function. Stem cell self-renewal/differentiation decisions must be carefully controlled during organogenesis, tissue homeostasis, and regeneration, as failure in stem cell maintenance or activation can lead to progressive tissue wasting, while unchecked self-renewal is a hallmark of many cancers. Here, we review evidence implicating the Notch signaling pathway, an evolutionarily conserved cell fate determinant with widespread roles in a variety of tissues and organisms, as a crucial regulator of stem cell behavior. As discussed below, this pathway plays varied and critical roles at multiple stages of organismal development, in lineage-specific differentiation of pluripotent embryonic stem cells, and in controlling stem cell numbers and activity in the context of age-related tissue degeneration, injury-induced tissue repair, and malignancy.
Collapse
|
45
|
Crews L, Rockenstein E, Masliah E. APP transgenic modeling of Alzheimer's disease: mechanisms of neurodegeneration and aberrant neurogenesis. Brain Struct Funct 2009; 214:111-26. [PMID: 20091183 PMCID: PMC2847155 DOI: 10.1007/s00429-009-0232-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 11/11/2009] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders of the aging population affect over 5 million people in the US and Europe alone. The common feature is the progressive accumulation of misfolded proteins with the formation of toxic oligomers. Alzheimer’s disease (AD) is characterized by cognitive impairment, progressive degeneration of neuronal populations in the neocortex and limbic system, and formation of amyloid plaques and neurofibrillary tangles. Amyloid-β (Aβ) is the product of proteolysis of amyloid precursor protein (APP) by β and γ-secretase enzymes. The neurodegenerative process in AD initiates with axonal and synaptic damage and is associated with progressive accumulation of toxic Aβ oligomers in the intracellular and extracellular space. In addition, neurodegeneration in AD is associated with alterations in neurogenesis. Aβ accumulation is the consequence of an altered balance between protein synthesis, aggregation rate, and clearance. Identification of genetic mutations in APP associated with familial forms of AD and gene polymorphisms associated with the more common sporadic variants of AD has led to the development of transgenic (tg) and knock out rodents as well as viral vector driven models of AD. While APP tg murine models with mutations in the N- and C-terminal flanking regions of Aβ are characterized by increased Aβ production with plaque formation, mutations in the mid-segment of Aβ result in increased formation of oligomers, and mutations toward the C-terminus (E22Q) segment results in amyloid angiopathy. Similar to AD, in APP tg models bearing familial mutations, formation of Aβ oligomers results in defective plasticity in the perforant pathway, selective neuronal degeneration, and alterations in neurogenesis. Promising results have been obtained utilizing APP tg models of AD to develop therapies including the use of β- and γ-secretase inhibitors, immunization, and stimulating neurogenesis.
Collapse
Affiliation(s)
- Leslie Crews
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | | | | |
Collapse
|