1
|
Li X, Wang G, Li W, Wang X, Wu J, He Y, Li X, Sun X, Zhang M, Guo Y. Histone deacetylase 9 plays a role in sevoflurane-induced neuronal differentiation inhibition by inactivating cAMP-response element binding protein transcription and inhibiting the expression of neurotrophin-3. FASEB J 2023; 37:e23164. [PMID: 37688590 DOI: 10.1096/fj.202300168r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/02/2023] [Accepted: 08/14/2023] [Indexed: 09/11/2023]
Abstract
Postoperative cognitive decline (POCD) is a common and serious complication following anesthesia and surgery; however, the precise mechanisms of POCD remain unclear. Our previous research showed that sevoflurane impairs adult hippocampal neurogenesis (AHN) and thus cognitive function in the aged brain by affecting neurotrophin-3 (NT-3) expression; however, the signaling mechanism involved remains unexplored. In this study, we found a dramatic decrease in the proportion of differentiated neurons with increasing concentrations of sevoflurane, and the inhibition of neural stem cell differentiation was partially reversed after the administration of exogenous NT-3. Understanding the molecular underpinnings by which sevoflurane affects NT-3 is key to counteracting cognitive dysfunction. Here, we report that sevoflurane administration for 2 days resulted in upregulation of histone deacetylase 9 (HDAC9) expression, which led to transcriptional inactivation of cAMP-response element binding protein (CREB). Due to the colocalization of HDAC9 and CREB within cells, this may be related to the interaction between HDAC9 and CREB. Anyway, this ultimately led to reduced NT-3 expression and inhibition of neural stem cell differentiation. Furthermore, knockdown of HDAC9 rescued the transcriptional activation of CREB after sevoflurane exposure, while reversing the downregulation of NT-3 expression and inhibition of neural stem cell differentiation. In summary, this study identifies a unique mechanism by which sevoflurane can inhibit CREB transcription through HDAC9, and this process reduces NT-3 levels and ultimately inhibits neuronal differentiation. This finding may reveal a new strategy to prevent sevoflurane-induced neuronal dysfunction.
Collapse
Affiliation(s)
- Xinlei Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Wei Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xu Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Jiangnan Wu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yingxue He
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xiaowei Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xiaobin Sun
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yanjing Guo
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
2
|
Firdoos S, Dai R, Tahir RA, Khan ZY, Li H, Zhang J, Ni J, Quan Z, Qing H. In silico identification of novel stilbenes analogs for potential multi-targeted drugs against Alzheimer's disease. J Mol Model 2023; 29:209. [PMID: 37314512 DOI: 10.1007/s00894-023-05609-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/27/2023] [Indexed: 06/15/2023]
Abstract
CONTEXT Alzheimer's disease (AD) is a chronic progressive neurodegenerative syndrome, which adversely disturbs cognitive abilities as well as intellectual processes and frequently occurs in the elderly. Inhibition of cholinesterase is a valuable approach to upsurge acetylcholine concentrations in the brain and persuades the development of multi-targeted ligands against cholinesterases. METHODS The current study aims to determine the binding potential accompanied by antioxidant and anti-inflammatory activities of stilbenes-designed analogs against both cholinesterases (Acetylcholinesterase and butyrylcholinesterase) and neurotrophin targets for effective AD therapeutics. Docking results have shown that the WS6 compound exhibited the least binding energy - 10.1 kcal/mol with Acetylcholinesterase and - 7.8 kcal/mol with butyrylcholinesterase. The WS6 also showed a better binding potential with neurotrophin targets that are Brain-derived Neurotrophic Factor, Neurotrophin 4, Nerve Growth Factor, and Neurotrophin 3. The tested compounds particularly WS6 revealed significant antioxidant and anti-inflammatory activities through the comparative docking analysis with Fluorouracil and Melatonin as control drugs of antioxidants while Celecoxib and Anakinra as anti-inflammatory. The bioinformatics approaches including molecular docking calculations followed by the pharmacokinetics analysis and molecular dynamic simulations were accomplished to explore the capabilities of designed stilbenes as effective and potential leads. Root mean square deviation, root mean square fluctuations, and MM-GBSA calculations were performed through molecular dynamic simulations to extract the structural and residual variations and binding free energies through the 50-ns time scale.
Collapse
Affiliation(s)
- Sundas Firdoos
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceutical, Beijing Institute of Technology (BIT), Beijing, 100081, People's Republic of China.
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceutical, Beijing Institute of Technology (BIT), Beijing, 100081, People's Republic of China.
| | - Rana Adnan Tahir
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Muzaffarabad, Pakistan
| | - Zahid Younas Khan
- Department of Computer Science and IT, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Jun Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
3
|
Zhao J, Huai J. Role of primary aging hallmarks in Alzheimer´s disease. Theranostics 2023; 13:197-230. [PMID: 36593969 PMCID: PMC9800733 DOI: 10.7150/thno.79535] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which severely threatens the health of the elderly and causes significant economic and social burdens. The causes of AD are complex and include heritable but mostly aging-related factors. The primary aging hallmarks include genomic instability, telomere wear, epigenetic changes, and loss of protein stability, which play a dominant role in the aging process. Although AD is closely associated with the aging process, the underlying mechanisms involved in AD pathogenesis have not been well characterized. This review summarizes the available literature about primary aging hallmarks and their roles in AD pathogenesis. By analyzing published literature, we attempted to uncover the possible mechanisms of aberrant epigenetic markers with related enzymes, transcription factors, and loss of proteostasis in AD. In particular, the importance of oxidative stress-induced DNA methylation and DNA methylation-directed histone modifications and proteostasis are highlighted. A molecular network of gene regulatory elements that undergoes a dynamic change with age may underlie age-dependent AD pathogenesis, and can be used as a new drug target to treat AD.
Collapse
|
4
|
Coradduzza D, Garroni G, Congiargiu A, Balzano F, Cruciani S, Sedda S, Nivoli A, Maioli M. MicroRNAs, Stem Cells in Bipolar Disorder, and Lithium Therapeutic Approach. Int J Mol Sci 2022; 23:ijms231810489. [PMID: 36142403 PMCID: PMC9502703 DOI: 10.3390/ijms231810489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
Bipolar disorder (BD) is a severe, chronic, and disabling neuropsychiatric disorder characterized by recurrent mood disturbances (mania/hypomania and depression, with or without mixed features) and a constellation of cognitive, psychomotor, autonomic, and endocrine abnormalities. The etiology of BD is multifactorial, including both biological and epigenetic factors. Recently, microRNAs (miRNAs), a class of epigenetic regulators of gene expression playing a central role in brain development and plasticity, have been related to several neuropsychiatric disorders, including BD. Moreover, an alteration in the number/distribution and differentiation potential of neural stem cells has also been described, significantly affecting brain homeostasis and neuroplasticity. This review aimed to evaluate the most reliable scientific evidence on miRNAs as biomarkers for the diagnosis of BD and assess their implications in response to mood stabilizers, such as lithium. Neural stem cell distribution, regulation, and dysfunction in the etiology of BD are also dissected.
Collapse
Affiliation(s)
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | | | - Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Stefania Sedda
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Alessandra Nivoli
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
- Correspondence: (A.N.); (M.M.); Tel.: +39-079-228-277 (A.N.); +39-079-255-406-228350 (M.M.)
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
- Correspondence: (A.N.); (M.M.); Tel.: +39-079-228-277 (A.N.); +39-079-255-406-228350 (M.M.)
| |
Collapse
|
5
|
Blanco-Luquin I, Acha B, Urdánoz-Casado A, Gómez-Orte E, Roldan M, Pérez-Rodríguez DR, Cabello J, Mendioroz M. NXN Gene Epigenetic Changes in an Adult Neurogenesis Model of Alzheimer's Disease. Cells 2022; 11:cells11071069. [PMID: 35406633 PMCID: PMC8998146 DOI: 10.3390/cells11071069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/17/2022] [Accepted: 03/20/2022] [Indexed: 12/10/2022] Open
Abstract
In view of the proven link between adult hippocampal neurogenesis (AHN) and learning and memory impairment, we generated a straightforward adult neurogenesis in vitro model to recapitulate DNA methylation marks in the context of Alzheimer’s disease (AD). Neural progenitor cells (NPCs) were differentiated for 29 days and Aβ peptide 1–42 was added. mRNA expression of Neuronal Differentiation 1 (NEUROD1), Neural Cell Adhesion Molecule 1 (NCAM1), Tubulin Beta 3 Class III (TUBB3), RNA Binding Fox-1 Homolog 3 (RBFOX3), Calbindin 1 (CALB1), and Glial Fibrillary Acidic Protein (GFAP) was determined by RT-qPCR to characterize the culture and framed within the multistep process of AHN. Hippocampal DNA methylation marks previously identified in Contactin-Associated Protein 1 (CNTNAP1), SEPT5-GP1BB Readthrough (SEPT5-GP1BB), T-Box Transcription Factor 5 (TBX5), and Nucleoredoxin (NXN) genes were profiled by bisulfite pyrosequencing or bisulfite cloning sequencing; mRNA expression was also measured. NXN outlined a peak of DNA methylation overlapping type 3 neuroblasts. Aβ-treated NPCs showed transient decreases of mRNA expression for SEPT5-GP1BB and NXN on day 9 or 19 and an increase in DNA methylation on day 29 for NXN. NXN and SEPT5-GP1BB may reflect alterations detected in the brain of AD human patients, broadening our understanding of this disease.
Collapse
Affiliation(s)
- Idoia Blanco-Luquin
- Neuroepigenetics Laboratory-Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA (Navarra Institute for Health Research), 31008 Pamplona, Spain; (B.A.); (A.U.-C.); (M.R.); (M.M.)
- Correspondence: ; Tel.: +34-848425739
| | - Blanca Acha
- Neuroepigenetics Laboratory-Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA (Navarra Institute for Health Research), 31008 Pamplona, Spain; (B.A.); (A.U.-C.); (M.R.); (M.M.)
| | - Amaya Urdánoz-Casado
- Neuroepigenetics Laboratory-Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA (Navarra Institute for Health Research), 31008 Pamplona, Spain; (B.A.); (A.U.-C.); (M.R.); (M.M.)
| | - Eva Gómez-Orte
- CIBIR (Center for Biomedical Research of La Rioja), 26006 Logroño, Spain; (E.G.-O.); (J.C.)
| | - Miren Roldan
- Neuroepigenetics Laboratory-Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA (Navarra Institute for Health Research), 31008 Pamplona, Spain; (B.A.); (A.U.-C.); (M.R.); (M.M.)
| | - Diego R. Pérez-Rodríguez
- Neurophysiology Department, Hospital Universitario de Navarra (HUN), IdiSNA (Navarra Institute for Health Research), 31008 Pamplona, Spain;
| | - Juan Cabello
- CIBIR (Center for Biomedical Research of La Rioja), 26006 Logroño, Spain; (E.G.-O.); (J.C.)
| | - Maite Mendioroz
- Neuroepigenetics Laboratory-Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA (Navarra Institute for Health Research), 31008 Pamplona, Spain; (B.A.); (A.U.-C.); (M.R.); (M.M.)
- Department of Neurology, Hospital Universitario de Navarra (HUN), IdiSNA (Navarra Institute for Health Research), 31008 Pamplona, Spain
| |
Collapse
|
6
|
Penning A, Tosoni G, Abiega O, Bielefeld P, Gasperini C, De Pietri Tonelli D, Fitzsimons CP, Salta E. Adult Neural Stem Cell Regulation by Small Non-coding RNAs: Physiological Significance and Pathological Implications. Front Cell Neurosci 2022; 15:781434. [PMID: 35058752 PMCID: PMC8764185 DOI: 10.3389/fncel.2021.781434] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/09/2021] [Indexed: 01/11/2023] Open
Abstract
The adult neurogenic niches are complex multicellular systems, receiving regulatory input from a multitude of intracellular, juxtacrine, and paracrine signals and biological pathways. Within the niches, adult neural stem cells (aNSCs) generate astrocytic and neuronal progeny, with the latter predominating in physiological conditions. The new neurons generated from this neurogenic process are functionally linked to memory, cognition, and mood regulation, while much less is known about the functional contribution of aNSC-derived newborn astrocytes and adult-born oligodendrocytes. Accumulating evidence suggests that the deregulation of aNSCs and their progeny can impact, or can be impacted by, aging and several brain pathologies, including neurodevelopmental and mood disorders, neurodegenerative diseases, and also by insults, such as epileptic seizures, stroke, or traumatic brain injury. Hence, understanding the regulatory underpinnings of aNSC activation, differentiation, and fate commitment could help identify novel therapeutic avenues for a series of pathological conditions. Over the last two decades, small non-coding RNAs (sncRNAs) have emerged as key regulators of NSC fate determination in the adult neurogenic niches. In this review, we synthesize prior knowledge on how sncRNAs, such as microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs), may impact NSC fate determination in the adult brain and we critically assess the functional significance of these events. We discuss the concepts that emerge from these examples and how they could be used to provide a framework for considering aNSC (de)regulation in the pathogenesis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Amber Penning
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Caterina Gasperini
- Neurobiology of miRNAs Lab, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Carlos P. Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
7
|
Wander CM, Song J. The neurogenic niche in Alzheimer's disease. Neurosci Lett 2021; 762:136109. [PMID: 34271133 PMCID: PMC9013442 DOI: 10.1016/j.neulet.2021.136109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/17/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022]
Abstract
Adult hippocampal neurogenesis is the process of generation and functional incorporation of new neurons, formed by adult neural stem cells in the dentate gyrus. Adult hippocampal neurogenesis is highly dependent upon the integration of dynamic external stimuli and is instrumental in the formation of new spatial memories. Adult hippocampal neurogenesis is therefore uniquely sensitive to the summation of neuronal circuit and neuroimmune environments that comprise the neurogenic niche, and has powerful implications in diseases of aging and neurological disorders. This sensitivity underlies the neurogenic niche alterations commonly observed in Alzheimer's disease, the most common form of dementia. This review summarizes Alzheimer's disease associated changes in neuronal network activity, neuroinflammatory processes, and adult neural stem cell fate choice that ultimately result in neurogenic niche dysfunction and impaired adult hippocampal neurogenesis. A more comprehensive understanding of the complex changes mediating neurogenic niche disturbances in Alzheimer's disease will aid development of future therapies targeting adult neurogenesis.
Collapse
Affiliation(s)
- Connor M Wander
- Department of Pharmacology, University of North Carolina at Chapel Hill
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Kahney EW, Zion EH, Sohn L, Viets-Layng K, Johnston R, Chen X. Characterization of histone inheritance patterns in the Drosophila female germline. EMBO Rep 2021; 22:e51530. [PMID: 34031963 PMCID: PMC8406404 DOI: 10.15252/embr.202051530] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 04/02/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022] Open
Abstract
Stem cells have the unique ability to undergo asymmetric division which produces two daughter cells that are genetically identical, but commit to different cell fates. The loss of this balanced asymmetric outcome can lead to many diseases, including cancer and tissue dystrophy. Understanding this tightly regulated process is crucial in developing methods to treat these abnormalities. Here, we report that during a Drosophila female germline stem cell asymmetric division, the two daughter cells differentially inherit histones at key genes related to either maintaining the stem cell state or promoting differentiation, but not at constitutively active or silenced genes. We combine histone labeling with DNA Oligopaints to distinguish old versus new histones and visualize their inheritance patterns at a single‐gene resolution in asymmetrically dividing cells in vivo. This strategy can be applied to other biological systems involving cell fate change during development or tissue homeostasis in multicellular organisms.
Collapse
Affiliation(s)
| | - Emily H Zion
- Department of Biology, The Johns Hopkins University, Baltimore, MD, USA
| | - Lydia Sohn
- Department of Biology, The Johns Hopkins University, Baltimore, MD, USA
| | - Kayla Viets-Layng
- Department of Biology, The Johns Hopkins University, Baltimore, MD, USA
| | - Robert Johnston
- Department of Biology, The Johns Hopkins University, Baltimore, MD, USA
| | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
9
|
Noureddini M, Bagheri-Mohammadi S. Adult Hippocampal Neurogenesis and Alzheimer's Disease: Novel Application of Mesenchymal Stem Cells and their Role in Hippocampal Neurogenesis. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:1-10. [PMID: 34268249 PMCID: PMC8256831 DOI: 10.22088/ijmcm.bums.10.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/07/2020] [Indexed: 10/31/2022]
Abstract
The neurogenesis can occur in two regions of the adult mammalian brain throughout the lifespan: the subgranular zone of the hippocampal dentate gyrus, and the subventricular zone of the lateral ventricle. The proliferation and maturation of neural progenitor cells are tightly regulated through intrinsic and extrinsic factors. The integration of maturated cells into the circuitry of the adult hippocampus emphasizes the importance of adult hippocampal neurogenesis in learning and memory. There is a large body of evidence demonstrating that alteration in the neurogenesis process in the adult hippocampus results in an early event in the course of Alzheimer's disease (AD). In AD condition, the number and maturation of neurons declines progressively in the hippocampus. Innovative therapies are required to modulate brain homeostasis. Mesenchymal stem cells (MSCs) hold an immense potential to regulate the neurogenesis process, and are currently tested in some brain-related disorders, such as AD. Therefore, the aim of this review is to discuss the use of MSCs to regulate endogenous adult neurogenesis and their significant impact on future strategies for the treatment of AD.
Collapse
Affiliation(s)
- Mahdi Noureddini
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeid Bagheri-Mohammadi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Interleukine-17 Administration Modulates Adult Hippocampal Neurogenesis and Improves Spatial Learning in Mice. J Mol Neurosci 2019; 69:254-263. [PMID: 31254254 DOI: 10.1007/s12031-019-01354-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/29/2019] [Indexed: 01/17/2023]
Abstract
Adult hippocampal neurogenesis plays an important role in health and disease. Regulating neurogenesis may be a key mechanism in the pathophysiology and treatment of several neurobehavioral disorders such as schizophrenia, depression, autism spectrum disorders and Alzheimer's disease. Cytokines are known to affect adult neurogenesis, but conflicting studies have been reported with regard to their actual role. Interleukine-17 (IL-17), a potent pro-inflammatory cytokine, has been shown to inhibit proliferation of neuroprogenitors and thus reduce hippocampal neurogenesis, while other studies suggested it can promote neurite outgrowth. In the present study we sought to explore the possible effect of a single dose administration of IL-17 on neurogenesis related behavior, i.e. spatial learning. Surprisingly, ICR mice injected with IL-17 (8 μg) had a significant slight improvement in spatial learning in the Morris water maze paradigm, without any changes in general locomotion compared with control mice. Indeed, the expression of neurogenesis related genes was down regulated following IL-17 treatment. However, we detected an upregulation in the expression of FGF-13, a gene promoting microtubule polymerization and neurite outgrowth, thus supporting neuronal maturation. We thus suggest that IL-17 has a complex role in regulating adult neurogenesis: inhibiting neuroprogenitors proliferation on one hand, while promoting maturation of already formed neuroblasts on the other hand. Our findings suggest that these roles can potentially affect neurogenesis related behavior. Its actual role in health and disease is yet to be determined.
Collapse
|
11
|
Chao F, Jiang L, Zhang Y, Zhou C, Zhang L, Tang J, Liang X, Qi Y, Zhu Y, Ma J, Tang Y. Stereological Investigation of the Effects of Treadmill Running Exercise on the Hippocampal Neurons in Middle-Aged APP/PS1 Transgenic Mice. J Alzheimers Dis 2019; 63:689-703. [PMID: 29689723 DOI: 10.3233/jad-171017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The risk of cognitive decline during Alzheimer's disease (AD) can be reduced if physical activity is maintained; however, the specific neural events underlying this beneficial effect are still uncertain. To quantitatively investigate the neural events underlying the effect of running exercise on middle-aged AD subjects, 12-month-old male APP/PS1 mice were randomly assigned to a control group or running group, and age-matched non-transgenic littermates were used as a wild-type group. AD running group mice were subjected to a treadmill running protocol (regular and moderate intensity) for four months. Spatial learning and memory abilities were assessed using the Morris water maze. Hippocampal amyloid plaques were observed using Thioflavin S staining and immunohistochemistry. Hippocampal volume, number of neurons, and number of newborn cells (BrdU+ cells) in the hippocampus were estimated using stereological techniques, and newborn neurons were observed using double-labelling immunofluorescence. Marked neuronal loss in both the CA1 field and dentate gyrus (DG) and deficits in both the neurogenesis and survival of new neurons in the DG of middle-aged APP/PS1 mice were observed. Running exercise could improve the spatial learning and memory abilities, reduce amyloid plaques in the hippocampi, delay neuronal loss, induce neurogenesis, and promote the survival of newborn neurons in the DG of middle-aged APP/PS1 mice. Exercise-induced protection of neurons and adult neurogenesis within the DG might be part of the important structural basis of the improved spatial learning and memory abilities observed in AD mice.
Collapse
Affiliation(s)
- Fenglei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Lin Jiang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yi Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Chunni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Xin Liang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yingqiang Qi
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yanqing Zhu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Ma
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
12
|
Zeng Q, Long Z, Feng M, Zhao Y, Luo S, Wang K, Wang Y, Yang G, He G. Valproic Acid Stimulates Hippocampal Neurogenesis via Activating the Wnt/β-Catenin Signaling Pathway in the APP/PS1/Nestin-GFP Triple Transgenic Mouse Model of Alzheimer's Disease. Front Aging Neurosci 2019; 11:62. [PMID: 30971911 PMCID: PMC6443965 DOI: 10.3389/fnagi.2019.00062] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/05/2019] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by the deposition of amyloid-β (Aβ) peptides and neurofibrillary tangles (NFTs) and massive loss of neuronal cells in the brain. Adult hippocampus continuously generates new neurons throughout life to shape brain function and impaired neurogenesis may contribute to a series of cognitive deterioration associated with AD. Enhancing endogenous neurogenesis represents a promising strategy that may ameliorate AD-associated cognitive defects. However, neurogenesis-enhancing approaches and underlying mechanisms are still not well studied. Here, using a mouse model of AD amyloid precursor protein (APP/PS1/Nestin-GFP triple transgenic mice, 3xTgAD), we examined the effects of 4 weeks of valproic acid (VPA) treatment on hippocampal neurogenesis in 2- and 6-month-old mice. VPA treatment promoted cell proliferation and increased the density of immature neurons in the dentate gyrus (DG) of the hippocampus of 3xTgAD mice. Consistent with enhanced neurogenesis, behavioral and morphological analysis showed that VPA treatment improved the learning and memory ability of 3xTgAD mice. Mechanistically, VPA treatment increased β-catenin levels, accumulated the inactive form of glycogen synthase kinase-3β (GSK-3β), and induced the expression of NeuroD1, a Wnt target gene involved in neurogenesis, suggesting the activation of the Wnt signaling pathway in the hippocampus of 3xTgAD mice. This study indicates that VPA stimulates neurogenesis in the adult hippocampus of AD mice model through the Wnt pathway, highlighting VPA as a potential therapeutic for treating AD and related diseases.
Collapse
Affiliation(s)
- Qinghua Zeng
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Zhimin Long
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Min Feng
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yueyang Zhao
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Shifang Luo
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Kejian Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Guang Yang
- Department of Medical Genetics, Cummings School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry and Molecular Biology, Cummings School of Medicine, University of Calgary, Calgary, AB, Canada.,Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Guiqiong He
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China.,Department of Anatomy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Leone L, Colussi C, Gironi K, Longo V, Fusco S, Li Puma DD, D'Ascenzo M, Grassi C. Altered Nup153 Expression Impairs the Function of Cultured Hippocampal Neural Stem Cells Isolated from a Mouse Model of Alzheimer's Disease. Mol Neurobiol 2019; 56:5934-5949. [PMID: 30689197 DOI: 10.1007/s12035-018-1466-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022]
Abstract
Impairment of adult hippocampal neurogenesis is an early event in Alzheimer's disease (AD), playing a crucial role in cognitive dysfunction associated with this pathology. However, the mechanisms underlying defective neurogenesis in AD are still unclear. Recently, the nucleoporin Nup153 has been described as a new epigenetic determinant of adult neural stem cell (NSC) maintenance and fate. Here we investigated whether Nup153 dysfunction could affect the plasticity of NSCs in AD. Nup153 expression was strongly reduced in AD-NSCs, as well as its interaction with the transcription factor Sox2, a master regulator of NSC stemness and their neuronal differentiation. Similar Nup153 reduction was also observed in WT-NSCs treated with amyloid-β (Aβ) or stimulated with a nitric oxide donor. Accordingly, AD-NSCs treated with either a γ-secretase inhibitor or antioxidant compounds showed higher Nup153 levels suggesting that both nitrosative stress and Aβ accumulation affect Nup153 expression. Of note, restoration of Nup153 levels in AD-NSCs promoted their proliferation, as assessed by BrdU incorporation, neurosphere assay, and stemness gene expression analysis. Nup153 overexpression also recovered AD-NSC response to differentiation, increasing the expression of pro-neuronal genes, the percentage of cells positive for neuronal markers, and the acquisition of a more mature neuronal phenotype. Electrophysiological recordings revealed that neurons differentiated from Nup153-transfected AD-NSCs displayed higher Na+ current density, comparable to those deriving from WT-NSCs. Our data uncover a novel role for Nup153 in NSCs from animal model of AD and point to Nup153 as potential target to restore physiological NSC behavior and fate in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lucia Leone
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Roma, Italia.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Claudia Colussi
- Institute of Cell Biology and Neurobiology, National Research Council, Largo F. Vito 1, 00168, Rome, Italy.
| | - Katia Gironi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Valentina Longo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Salvatore Fusco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Roma, Italia.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Domenica Donatella Li Puma
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Roma, Italia.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Marcello D'Ascenzo
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Roma, Italia.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Roma, Italia.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| |
Collapse
|
14
|
Abstract
All people want to age "successfully," maintaining functional capacity and quality of life as they reach advanced age. Achieving this goal depends on preserving optimal cognitive and brain functioning. Yet, significant individual differences exist in this regard. Some older adults continue to retain most cognitive abilities throughout their lifetime. Others experience declines in cognitive and functional capacity that range from mild decrements in certain cognitive functions over time to severe dementia among those with neurodegenerative diseases. Even among relatively healthy "successful agers," certain cognitive functions are reduced from earlier levels. This is particularly true for cognitive functions that are dependent on cognitive processing speed and efficiency. Working memory and executive and attentional functions tend to be most vulnerable. Learning and memory functions are also usually reduced, although in the absence of neurodegenerative disease learning and retrieval efficiency rather than memory storage are affected. Other functions, such as visual perception, language, semantics, and knowledge, are often well preserved. Structural, functional, and physiologic/metabolic brain changes correspond with age-associated cognitive decline. Physiologic and metabolic mechanisms, such as oxidative stress and neuroinflammation, may contribute to these changes, along with the contribution of comorbidities that secondarily affect the brain of older adults. Cognitive frailty often corresponds with physical frailty, both affected by multiple exogenous and endogenous factors. Neuropsychologic assessment provides a way of measuring the cognitive and functional status of older adults, which is useful for monitoring changes that may be occurring. Neuroimaging is also useful for characterizing age-associated structural, functional, physiologic, and metabolic brain changes, including alterations in cerebral blood flow and metabolite concentrations. Some interventions that may enhance cognitive function, such as cognitive training, neuromodulation, and pharmacologic approaches, exist or are being developed. Yet, preventing, slowing, and reversing the adverse effects of cognitive aging remains a challenge.
Collapse
Affiliation(s)
- Ronald A Cohen
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States; Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States.
| | - Michael M Marsiske
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States; Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States
| | - Glenn E Smith
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States; Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Liu Y, Xu J, Zhu F, Ye H, Hu C, Huang J, Zheng Y. Research advances in the regulation of the putative ovarian germline stem cell niche on female germline stem cells. Syst Biol Reprod Med 2018; 65:121-128. [PMID: 30204491 DOI: 10.1080/19396368.2018.1515272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Stem cells are ideal seeding cells, which have the potential for self-renewal and multiple differentiation, and they play a fundamental role in maintaining homeostasis and regenerating and repairing tissue. The discovery of female germline stem cells (FGSCs) brings much hope for the postnatal renewal of oocytes and solving some female infertility problems. Ovarian function declines with increasing female age. Moreover, ovarian germline stem cell niche-aging could be the main cause of ovarian senescence, which ultimately leads to decreased follicle generation, declining female fertility, and age-related diseases, such as osteoporosis and ovarian cancer. The ovarian germline stem cell niche is the surrounding microenvironment in which FGSCs live, and it helps control the biological characteristics of FGSCs in many ways, such as nutritional supply and immunological cytokine secretion. This paper reviews the knowledge about the ovarian germline stem cell niche and its probable regulatory mechanisms on FGSCs, which provides valuable scientific information and scope for the prevention and treatment of ovarian senescence. Abbreviations: BMP: bone morphogenetic protein; Dpp: decapentaplegic; FGSC: female germline stem cell; IL, interleukin; OGSC: ovarian germline stem cells; ROS: reactive oxygen species; TGF, transforming growth factor; TNF, tumor necrosis factor.
Collapse
Affiliation(s)
- Yangchun Liu
- a Jiangxi Medical College , Nanchang University , Nanchang , Jiangxi , PR China.,b Queen Mary College of Nanchang University , Nanchang , Jiangxi , PR China
| | - Jiao Xu
- a Jiangxi Medical College , Nanchang University , Nanchang , Jiangxi , PR China.,c First Clinical College of Nanchang University , Nanchang , Jiangxi , PR China
| | - Feiyin Zhu
- a Jiangxi Medical College , Nanchang University , Nanchang , Jiangxi , PR China.,b Queen Mary College of Nanchang University , Nanchang , Jiangxi , PR China
| | - Haifeng Ye
- a Jiangxi Medical College , Nanchang University , Nanchang , Jiangxi , PR China.,d The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province , Nanchang , Jiangxi , PR China
| | - Chuan Hu
- a Jiangxi Medical College , Nanchang University , Nanchang , Jiangxi , PR China.,d The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province , Nanchang , Jiangxi , PR China
| | - Jian Huang
- a Jiangxi Medical College , Nanchang University , Nanchang , Jiangxi , PR China.,d The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province , Nanchang , Jiangxi , PR China
| | - Yuehui Zheng
- a Jiangxi Medical College , Nanchang University , Nanchang , Jiangxi , PR China.,d The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province , Nanchang , Jiangxi , PR China
| |
Collapse
|
16
|
Directing neuronal cell fate in vitro : Achievements and challenges. Prog Neurobiol 2018; 168:42-68. [DOI: 10.1016/j.pneurobio.2018.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/30/2018] [Accepted: 04/05/2018] [Indexed: 12/22/2022]
|
17
|
Kulminski AM, Huang J, Wang J, He L, Loika Y, Culminskaya I. Apolipoprotein E region molecular signatures of Alzheimer's disease. Aging Cell 2018; 17:e12779. [PMID: 29797398 PMCID: PMC6052488 DOI: 10.1111/acel.12779] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2018] [Indexed: 01/01/2023] Open
Abstract
Although the APOE region is the strongest genetic risk factor for Alzheimer's diseases (ADs), its pathogenic role remains poorly understood. Elucidating genetic predisposition to ADs, a subset of age-related diseases characteristic for postreproductive period, is hampered by the undefined role of evolution in establishing molecular mechanisms of such diseases. This uncertainty is inevitable source of natural-selection-free genetic heterogeneity in predisposition to ADs. We performed first large-scale analysis of linkage disequilibrium (LD) structures characterized by 30 polymorphisms from five genes in the APOE 19q13.3 region (BCAM, NECTIN2, TOMM40, APOE, and APOC1) in 2,673 AD-affected and 16,246 unaffected individuals from five cohorts. Consistent with the undefined role of evolution in age-related diseases, we found that these structures, being highly heterogeneous, are significantly different in subjects with and without ADs. The pattern of the difference represents molecular signature of AD comprised of single nucleotide polymorphisms (SNPs) from all five genes in the APOE region. Significant differences in LD in subjects with and without ADs indicate SNPs from different genes likely involved in AD pathogenesis. Significant and highly heterogeneous molecular signatures of ADs provide unprecedented insight into complex polygenetic predisposition to ADs in the APOE region. These findings are more consistent with a complex haplotype than with a single genetic variant origin of ADs in this region.
Collapse
Affiliation(s)
- Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Jian Huang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Jiayi Wang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| |
Collapse
|
18
|
Fluoxetine attenuates the impairment of spatial learning ability and prevents neuron loss in middle-aged APPswe/PSEN1dE9 double transgenic Alzheimer's disease mice. Oncotarget 2018; 8:27676-27692. [PMID: 28430602 PMCID: PMC5438600 DOI: 10.18632/oncotarget.15398] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 01/31/2017] [Indexed: 01/04/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) have been reported to increase cognitive performance in some clinical studies of Alzheimer's disease (AD). However, there is a lack of evidence supporting the efficacy of SSRIs as cognition enhancers in AD, and the role of SSRIs as a treatment for AD remains largely unclear. Here, we characterized the impact of fluoxetine (FLX), a well-known SSRI, on neurons in the dentate gyrus (DG) and in CA1 and CA3 of the hippocampus of middle-aged (16 to 17 months old) APPswe/PSEN1dE9 (APP/PS1) transgenic AD model mice. We found that intraperitoneal (i.p.) injection of FLX (10 mg/kg/day) for 5 weeks effectively alleviated the impairment of spatial learning ability in middle-aged APP/PS1 mice as evaluated using the Morris water maze. More importantly, the number of neurons in the hippocampal DG was significantly increased by FLX. Additionally, FLX reduced the deposition of beta amyloid, inhibited GSK-3β activity and increased the level of β-catenin in middle-aged APP/PS1 mice. Collectively, the results of this study indicate that FLX delayed the progression of neuronal loss in the hippocampal DG in middle-aged AD mice, and this effect may underlie the FLX-induced improvement in learning ability. FLX may therefore serve as a promising therapeutic drug for AD.
Collapse
|
19
|
Chen JJ, Wang T, An CD, Jiang CY, Zhao J, Li S. Brain-derived neurotrophic factor: a mediator of inflammation-associated neurogenesis in Alzheimer's disease. Rev Neurosci 2018; 27:793-811. [PMID: 27508959 DOI: 10.1515/revneuro-2016-0017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/29/2016] [Indexed: 12/11/2022]
Abstract
In early- or late-onset Alzheimer's disease (AD), inflammation, which is triggered by pathologic conditions, influences the progression of neurodegeneration. Brain-derived neurotrophic factor (BDNF) has emerged as a crucial mediator of neurogenesis, because it exhibits a remarkable activity-dependent regulation of expression, which suggests that it may link inflammation to neurogenesis. Emerging evidence suggests that acute and chronic inflammation in AD differentially modulates neurotrophin functions, which are related to the roles of inflammation in neuroprotection and neurodegeneration. Recent studies also indicate novel mechanisms of BDNF-mediated neuroprotection, including the modulation of autophagy. Numerous research studies have demonstrated reverse parallel alterations between proinflammatory cytokines and BDNF during neurodegeneration; thus, we hypothesize that one mechanism that underlies the negative impact of chronic inflammation on neurogenesis is the reduction of BDNF production and function by proinflammatory cytokines.
Collapse
|
20
|
Abstract
The role of DNA methylation in brain development is an intense area of research because the brain has particularly high levels of CpG and mutations in many of the proteins involved in the establishment, maintenance, interpretation, and removal of DNA methylation impact brain development and/or function. These include DNA methyltransferase (DNMT), Ten-Eleven Translocation (TET), and Methyl-CpG binding proteins (MBPs). Recent advances in sequencing breadth and depth as well the detection of different forms of methylation have greatly expanded our understanding of the diversity of DNA methylation in the brain. The contributions of DNA methylation and associated proteins to embryonic and adult neurogenesis will be examined. Particular attention will be given to the impact on adult hippocampal neurogenesis (AHN), which is a key mechanism contributing to brain plasticity, learning, memory and mood regulation. DNA methylation influences multiple aspects of neurogenesis from stem cell maintenance and proliferation, fate specification, neuronal differentiation and maturation, and synaptogenesis. In addition, DNA methylation during neurogenesis has been shown to be responsive to many extrinsic signals, both under normal conditions and during disease and injury. Finally, crosstalk between DNA methylation, Methyl-DNA binding domain (MBD) proteins such as MeCP2 and MBD1 and histone modifying complexes is used as an example to illustrate the extensive interconnection between these epigenetic regulatory systems.
Collapse
Affiliation(s)
- Emily M Jobe
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.,Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Xinyu Zhao
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.,Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
21
|
Shi Z, Hong Y, Zhang K, Wang J, Zheng L, Zhang Z, Hu Z, Han X, Han Y, Chen T, Yao Q, Cui H, Hong W. BAG-1M co-activates BACE1 transcription through NF-κB and accelerates Aβ production and memory deficit in Alzheimer's disease mouse model. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2398-2407. [PMID: 28502705 DOI: 10.1016/j.bbadis.2017.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 05/01/2017] [Accepted: 05/10/2017] [Indexed: 12/13/2022]
Abstract
Accumulation of amyloid β protein (Aβ)-containing neuritic plaques in the brain is a neuropathological feature of Alzheimer's disease (AD). The β-site APP-cleaving enzyme 1 (BACE1) is essential for Aβ generation and dysregulation of BACE1 expression may lead to AD pathogenesis. Bcl-2-associated athanogen 1M (BAG-1M), initially identified as an anti-apoptotic protein, has also been found to be highly expressed in the same neurons that contain intracellular amyloid in the hippocampus of AD patient. In this report, we found that over-expression of BAG-1M enhances BACE1-mediated cleavage of amyloid precursor protein (APP) and Aβ production by up-regulating BACE1 gene transcription. The regulation of BACE1 transcription by BAG-1M was dependent on NF-κB, as BAG-1M complexes NF-κB at the promoter of BACE1 gene and co-activates NF-κB-facilitated BACE1 transcription. Moreover, expression of BAG-1M by lentiviral vector in the hippocampus of AD transgenic model mice promotes Aβ generation and formation of neuritic plaque, and subsequently accelerates memory deficits of the mice. These results provide evidence for an emerging role of BAG-1M in the regulation of BACE1 expression and AD pathogenesis and that targeting the BAG-1M-NF-κB complex may provide a mechanism for inhibiting Aβ production and plaque formation.
Collapse
Affiliation(s)
- Zhemin Shi
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yuheng Hong
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Kun Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jingzhao Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lina Zheng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zhen Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zhimei Hu
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaohui Han
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yawei Han
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Ting Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Qingbin Yao
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hongmei Cui
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Wei Hong
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
22
|
Yokoyama AS, Rutledge JC, Medici V. DNA methylation alterations in Alzheimer's disease. ENVIRONMENTAL EPIGENETICS 2017; 3:dvx008. [PMID: 29492310 PMCID: PMC5804548 DOI: 10.1093/eep/dvx008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/22/2017] [Accepted: 03/22/2017] [Indexed: 05/24/2023]
Abstract
The observation that Alzheimer's disease (AD) patients with similar and even identical genetic backgrounds often present with heterogeneous pathologies has prompted the hypothesis that epigenetics may contribute to AD. While the study of epigenetics encompasses a variety of modifications including histone modifications and non-coding RNAs, much of the research on how epigenetics might impact AD pathology has been focused on DNA methylation. To this end, several studies have characterized DNA methylation alterations in various brain regions of individuals with AD, with conflicting results. This review examines the results of studies analyzing both global and gene-specific DNA methylation changes in AD and also assesses the results of studies analyzing DNA hydroxymethylation in patients with AD.
Collapse
Affiliation(s)
- Amy S. Yokoyama
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| | - John C. Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| | - Valentina Medici
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| |
Collapse
|
23
|
SETD4 Regulates Cell Quiescence and Catalyzes the Trimethylation of H4K20 during Diapause Formation in Artemia. Mol Cell Biol 2017; 37:MCB.00453-16. [PMID: 28031330 DOI: 10.1128/mcb.00453-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/02/2016] [Indexed: 01/19/2023] Open
Abstract
As a prominent characteristic of cell life, the regulation of cell quiescence is important for proper development, regeneration, and stress resistance and may play a role in certain degenerative diseases. However, the mechanism underlying quiescence remains largely unknown. Encysted embryos of Artemia are useful for studying the regulation of this state because they remain quiescent for prolonged periods during diapause, a state of obligate dormancy. In the present study, SET domain-containing protein 4, a histone lysine methyltransferase from Artemia, was identified, characterized, and named Ar-SETD4. We found that Ar-SETD4 was expressed abundantly in Artemia diapause embryos, in which cells were in a quiescent state. Meanwhile, trimethylated histone H4K20 (H4K20me3) was enriched in diapause embryos. The knockdown of Ar-SETD4 reduced the level of H4K20me3 significantly and prevented the formation of diapause embryos in which neither the cell cycle nor embryogenesis ceased. The catalytic activity of Ar-SETD4 on H4K20me3 was confirmed by an in vitro histone methyltransferase (HMT) assay and overexpression in cell lines. This study provides insights into the function of SETD4 and the mechanism of cell quiescence regulation.
Collapse
|
24
|
Shukla M, Govitrapong P, Boontem P, Reiter RJ, Satayavivad J. Mechanisms of Melatonin in Alleviating Alzheimer's Disease. Curr Neuropharmacol 2017; 15:1010-1031. [PMID: 28294066 PMCID: PMC5652010 DOI: 10.2174/1570159x15666170313123454] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/10/2017] [Accepted: 03/09/2017] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic, progressive and prevalent neurodegenerative disease characterized by the loss of higher cognitive functions and an associated loss of memory. The thus far "incurable" stigma for AD prevails because of variations in the success rates of different treatment protocols in animal and human studies. Among the classical hypotheses explaining AD pathogenesis, the amyloid hypothesis is currently being targeted for drug development. The underlying concept is to prevent the formation of these neurotoxic peptides which play a central role in AD pathology and trigger a multispectral cascade of neurodegenerative processes post-aggregation. This could possibly be achieved by pharmacological inhibition of β- or γ-secretase or stimulating the nonamyloidogenic α-secretase. Melatonin the pineal hormone is a multifunctioning indoleamine. Production of this amphiphilic molecule diminishes with advancing age and this decrease runs parallel with the progression of AD which itself explains the potential benefits of melatonin in line of development and devastating consequences of the disease progression. Our recent studies have revealed a novel mechanism by which melatonin stimulates the nonamyloidogenic processing and inhibits the amyloidogenic processing of β-amyloid precursor protein (βAPP) by stimulating α -secretases and consequently down regulating both β- and γ-secretases at the transcriptional level. In this review, we discuss and evaluate the neuroprotective functions of melatonin in AD pathogenesis, including its role in the classical hypotheses in cellular and animal models and clinical interventions in AD patients, and suggest that with early detection, melatonin treatment is qualified to be an anti-AD therapy.
Collapse
Affiliation(s)
- Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 54 Kamphaeng Phet 6 Road, Lak Si, Bangkok10210, Thailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 54 Kamphaeng Phet 6 Road, Lak Si, Bangkok10210, Thailand
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Parichart Boontem
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 54 Kamphaeng Phet 6 Road, Lak Si, Bangkok10210, Thailand
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jutamaad Satayavivad
- Chulabhorn Research Institute and Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok10210, Thailand
| |
Collapse
|
25
|
Horgusluoglu E, Nudelman K, Nho K, Saykin AJ. Adult neurogenesis and neurodegenerative diseases: A systems biology perspective. Am J Med Genet B Neuropsychiatr Genet 2017; 174:93-112. [PMID: 26879907 PMCID: PMC4987273 DOI: 10.1002/ajmg.b.32429] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/29/2016] [Indexed: 12/21/2022]
Abstract
New neurons are generated throughout adulthood in two regions of the brain, the olfactory bulb and dentate gyrus of the hippocampus, and are incorporated into the hippocampal network circuitry; disruption of this process has been postulated to contribute to neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. Known modulators of adult neurogenesis include signal transduction pathways, the vascular and immune systems, metabolic factors, and epigenetic regulation. Multiple intrinsic and extrinsic factors such as neurotrophic factors, transcription factors, and cell cycle regulators control neural stem cell proliferation, maintenance in the adult neurogenic niche, and differentiation into mature neurons; these factors act in networks of signaling molecules that influence each other during construction and maintenance of neural circuits, and in turn contribute to learning and memory. The immune system and vascular system are necessary for neuronal formation and neural stem cell fate determination. Inflammatory cytokines regulate adult neurogenesis in response to immune system activation, whereas the vasculature regulates the neural stem cell niche. Vasculature, immune/support cell populations (microglia/astrocytes), adhesion molecules, growth factors, and the extracellular matrix also provide a homing environment for neural stem cells. Epigenetic changes during hippocampal neurogenesis also impact memory and learning. Some genetic variations in neurogenesis related genes may play important roles in the alteration of neural stem cells differentiation into new born neurons during adult neurogenesis, with important therapeutic implications. In this review, we discuss mechanisms of and interactions between these modulators of adult neurogenesis, as well as implications for neurodegenerative disease and current therapeutic research. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Emrin Horgusluoglu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kelly Nudelman
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Andrew J. Saykin
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
26
|
Reddy RG, Veeraval L, Maitra S, Chollet-Krugler M, Tomasi S, Dévéhat FLL, Boustie J, Chakravarty S. Lichen-derived compounds show potential for central nervous system therapeutics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1527-1534. [PMID: 27765373 DOI: 10.1016/j.phymed.2016.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 08/16/2016] [Accepted: 08/27/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Natural products from lichens are widely investigated for their biological properties, yet their potential as central nervous system (CNS) therapeutic agents is less explored. PURPOSE The present study investigated the neuroactive properties of selected lichen compounds (atranorin, perlatolic acid, physodic acid and usnic acid), for their neurotrophic, neurogenic and acetylcholine esterase (AChE) activities. METHODS Neurotrophic activity (neurite outgrowth) was determined using murine neuroblastoma Neuro2A cells. A MTT assay was performed to assess the cytotoxicity of compounds at optimum neurotrophic activity. Neuro2A cells treated with neurotrophic lichen compounds were used for RT-PCR to evaluate the induction of genes that code for the neurotrophic markers BDNF and NGF. Immunoblotting was used to assess acetyl H3 and H4 levels, the epigenetic markers associated with neurotrophic and/or neurogenic activity. The neurogenic property of the compounds was determined using murine hippocampal primary cultures. AChE inhibition activity was performed using a modified Ellman's esterase method. RESULTS Lichen compounds atranorin, perlatolic acid, physodic acid and (+)-usnic acid showed neurotrophic activity in a preliminary cell-based screening based on Neuro2A neurite outgrowth. Except for usnic acid, no cytotoxic effects were observed for the two depsides (atranorin and perlatolic acid) and the alkyl depsidone (physodic acid). Perlatolic acid appears to be promising, as it also exhibited AChE inhibition activity and potent proneurogenic activity. The neurotrophic lichen compounds (atranorin, perlatolic acid, physodic acid) modulated the gene expression of BDNF and NGF. In addition, perlatolic acid showed increased protein levels of acetyl H3 and H4 in Neuro2A cells. CONCLUSION These lichen depsides and depsidones showed neuroactive properties in vitro (Neuro2A cells) and ex vivo (primary neural stem or progenitor cells), suggesting their potential to treat CNS disorders.
Collapse
Affiliation(s)
- R Gajendra Reddy
- Chemical Biology, CSIR- Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Lenin Veeraval
- Chemical Biology, CSIR- Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Swati Maitra
- Chemical Biology, CSIR- Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Marylène Chollet-Krugler
- PNSCM-UMRCNRS 6226, ISCR, Faculté des Sciences Pharmaceutiques et Biologiques, Université Européenne de Bretagne, Université de Rennes 1, 2 Av. du Pr Léon Bernard, 35043 Rennes Cedex, France
| | - Sophie Tomasi
- PNSCM-UMRCNRS 6226, ISCR, Faculté des Sciences Pharmaceutiques et Biologiques, Université Européenne de Bretagne, Université de Rennes 1, 2 Av. du Pr Léon Bernard, 35043 Rennes Cedex, France
| | - Françoise Lohézic-Le Dévéhat
- PNSCM-UMRCNRS 6226, ISCR, Faculté des Sciences Pharmaceutiques et Biologiques, Université Européenne de Bretagne, Université de Rennes 1, 2 Av. du Pr Léon Bernard, 35043 Rennes Cedex, France
| | - Joël Boustie
- PNSCM-UMRCNRS 6226, ISCR, Faculté des Sciences Pharmaceutiques et Biologiques, Université Européenne de Bretagne, Université de Rennes 1, 2 Av. du Pr Léon Bernard, 35043 Rennes Cedex, France.
| | - Sumana Chakravarty
- Chemical Biology, CSIR- Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India.
| |
Collapse
|
27
|
de Bem CMBE, Pezzi JC, Borba EM, Chaves MLF, de Andrade FM, Fiegenbaum M, Camozzato A. The synergistic risk effect of apolipoprotein ε4 and DNA (cytosine-5-)-methyltransferase 3 beta (DNMT3B) haplotype for Alzheimer's disease. Mol Biol Rep 2016; 43:653-8. [PMID: 27188425 DOI: 10.1007/s11033-016-3999-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 05/04/2016] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is a complex and multifactorial disease with the contribution of several genes and polymorphisms to its development. Among these genes, the APOEε4 is the best known risk factor for AD. Methylation is associated with APOE expression and AD development. Recently, we found an association of the TGG haplotype in the DNMT3B gene, one of the catalyst enzyme for methylation, with AD. Therefore, the objective of the study was to investigate whether APOEε4 and TGG haplotype have an synergistic effect on AD. The sample was composed of 212 Caucasian individuals (108 healthy controls and 104 with AD by NINCDS-ADRDA and DSM-IV-TR criteria) from southern Brazil. The genetic analyses were performed by real time PCR for TaqMan(®) assay. Multivariate logistic regression was performed categorizing groups according to presence of APOEε4 and/or TGG haplotype as an independent variable for outcome AD. The presence of TGG haplotype plus the allele APOEε4 were strongly associated with AD [OR 11.13; 95 % CI (4.25-29.16); P < 0.001]. This association had a higher risk than each risk factor alone. We found a strong association of the interaction of DNMT3B gene with the APOEε4 in this sample of AD patients. The presence of TGG haplotype and APOEε4 significantly increased the risk of developing the disease, showing an synergistic effect.
Collapse
Affiliation(s)
- Cíntia Monique Boschmann Ens de Bem
- Post Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua: Taquara 564/306 Bairro Petrópolis, Porto Alegre, RS, 90460210, Brazil
| | - Julio Carlos Pezzi
- Post Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua: Taquara 564/306 Bairro Petrópolis, Porto Alegre, RS, 90460210, Brazil
| | - Ericksen Mielle Borba
- Neurology Service, Dementia Clinic, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | | | | | - Marilu Fiegenbaum
- Post Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua: Taquara 564/306 Bairro Petrópolis, Porto Alegre, RS, 90460210, Brazil
| | - Analuiza Camozzato
- Post Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua: Taquara 564/306 Bairro Petrópolis, Porto Alegre, RS, 90460210, Brazil.
| |
Collapse
|
28
|
Liu H, Song N. Molecular Mechanism of Adult Neurogenesis and its Association with Human Brain Diseases. J Cent Nerv Syst Dis 2016; 8:5-11. [PMID: 27375363 PMCID: PMC4915785 DOI: 10.4137/jcnsd.s32204] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 04/07/2016] [Accepted: 05/30/2016] [Indexed: 12/19/2022] Open
Abstract
Recent advances in neuroscience challenge the old dogma that neurogenesis occurs only during embryonic development. Mounting evidence suggests that functional neurogenesis occurs throughout adulthood. This review article discusses molecular factors that affect adult neurogenesis, including morphogens, growth factors, neurotransmitters, transcription factors, and epigenetic factors. Furthermore, we summarize and compare current evidence of associations between adult neurogenesis and human brain diseases such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and brain tumors.
Collapse
Affiliation(s)
- He Liu
- Department of Biology, Morosky College of Health Professions and Sciences, Gannon University, Erie, PA, USA
| | - Ni Song
- Division of Health Sciences & Workforce Technology, Lamar State College-Orange, Orange, TX, USA
| |
Collapse
|
29
|
Li X, Bao X, Wang R. Neurogenesis-based epigenetic therapeutics for Alzheimer's disease (Review). Mol Med Rep 2016; 14:1043-53. [DOI: 10.3892/mmr.2016.5390] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 04/14/2016] [Indexed: 11/06/2022] Open
|
30
|
Mastroeni D, Chouliaras L, Van den Hove DL, Nolz J, Rutten BP, Delvaux E, Coleman. PD. Increased 5-hydroxymethylation levels in the sub ventricular zone of the Alzheimer's brain. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.nepig.2016.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
31
|
Herbert J, Lucassen PJ. Depression as a risk factor for Alzheimer's disease: Genes, steroids, cytokines and neurogenesis - What do we need to know? Front Neuroendocrinol 2016; 41:153-71. [PMID: 26746105 DOI: 10.1016/j.yfrne.2015.12.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/23/2015] [Accepted: 12/27/2015] [Indexed: 01/18/2023]
Abstract
Depression (MDD) is prodromal to, and a component of, Alzheimer's disease (AD): it may also be a trigger for incipient AD. MDD is not a unitary disorder, so there may be particular subtypes of early life MDD that pose independent high risks for later AD, though the identification of these subtypes is problematical. There may either be a common pathological event underlying both MDD and AD, or MDD may sensitize the brain to a second event ('hit') that precipitates AD. MDD may also accelerate brain ageing, including altered DNA methylation, increased cortisol but decreasing DHEA and thus the risk for AD. So far, genes predicting AD (e.g. APOEε4) are not risk factors for MDD, and those implicated in MDD (e.g. SLC6A4) are not risks for AD, so a common genetic predisposition looks unlikely. There is as yet no strong indication that an epigenetic event occurs during some forms of MDD that predisposes to later AD, though the evidence is limited. Glucocorticoids (GCs) are disturbed in some cases of MDD and in AD. GCs have marked degenerative actions on the hippocampus, a site of early β-amyloid deposition, and rare genetic variants of GC-regulating enzymes (e.g. 11β-HSD) predispose to AD. GCs also inhibit hippocampal neurogenesis and plasticity, and thus episodic memory, a core symptom of AD. Disordered GCs in MDD may inhibit neurogenesis, but the contribution of diminished neurogenesis to the onset or progression of AD is still debated. GCs and cytokines also reduce BDNF, implicated in both MDD and AD and hippocampal neurogenesis, reinforcing the notion that those cases of MDD with disordered GCs may be a risk for AD. Cytokines, including IL1β, IL6 and TNFα, are increased in the blood in some cases of MDD. They also reduce hippocampal neurogenesis, and increased cytokines are a known risk for later AD. Inflammatory changes occur in both MDD and AD (e.g. raised CRP, TNFα). Both cytokines and GCs can have pro-inflammatory actions in the brain. Inflammation (e.g. microglial activation) may be a common link, but this has not been systematically investigated. We lack substantial, rigorous and comprehensive follow-up studies to better identify possible subtypes of MDD that may represent a major predictor for later AD. This would enable specific interventions during critical episodes of these subtypes of MDD that should reduce this substantial risk.
Collapse
Affiliation(s)
- Joe Herbert
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, UK.
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands
| |
Collapse
|
32
|
Kinjo ER, Higa GSV, Santos BA, de Sousa E, Damico MV, Walter LT, Morya E, Valle AC, Britto LRG, Kihara AH. Pilocarpine-induced seizures trigger differential regulation of microRNA-stability related genes in rat hippocampal neurons. Sci Rep 2016; 6:20969. [PMID: 26869208 PMCID: PMC4751485 DOI: 10.1038/srep20969] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 01/14/2016] [Indexed: 11/17/2022] Open
Abstract
Epileptogenesis in the temporal lobe elicits regulation of gene expression and protein translation, leading to reorganization of neuronal networks. In this process, miRNAs were described as being regulated in a cell-specific manner, although mechanistics of miRNAs activity are poorly understood. The specificity of miRNAs on their target genes depends on their intracellular concentration, reflecting the balance of biosynthesis and degradation. Herein, we confirmed that pilocarpine application promptly (<30 min) induces status epilepticus (SE) as revealed by changes in rat electrocorticogram particularly in fast-beta range (21–30 Hz). SE simultaneously upregulated XRN2 and downregulated PAPD4 gene expression in the hippocampus, two genes related to miRNA degradation and stability, respectively. Moreover, SE decreased the number of XRN2-positive cells in the hilus, while reduced the number of PAPD4-positive cells in CA1. XRN2 and PAPD4 levels did not change in calretinin- and CamKII-positive cells, although it was possible to determine that PAPD4, but not XRN2, was upregulated in parvalbumin-positive cells, revealing that SE induction unbalances the accumulation of these functional-opposed proteins in inhibitory interneurons that directly innervate distinct domains of pyramidal cells. Therefore, we were able to disclose a possible mechanism underlying the differential regulation of miRNAs in specific neurons during epileptogenesis.
Collapse
Affiliation(s)
- Erika R Kinjo
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Guilherme S V Higa
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil.,Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Bianca A Santos
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Erica de Sousa
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Marcio V Damico
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Lais T Walter
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Edgard Morya
- Programa de Neuroengenharia, Instituto Internacional de Neurociências Edmond e Lily Safra, ISD, Macaíba, RN, Brazil
| | - Angela C Valle
- Laboratório de Neurociências, LIM 01, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Luiz R G Britto
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Alexandre H Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil.,Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
33
|
Role of Epigenetics in Stem Cell Proliferation and Differentiation: Implications for Treating Neurodegenerative Diseases. Int J Mol Sci 2016; 17:ijms17020199. [PMID: 26848657 PMCID: PMC4783933 DOI: 10.3390/ijms17020199] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 01/17/2016] [Accepted: 01/27/2016] [Indexed: 12/15/2022] Open
Abstract
The main objectives of this review are to survey the current literature on the role of epigenetics in determining the fate of stem cells and to assess how this information can be used to enhance the treatment strategies for some neurodegenerative disorders, like Huntington’s disease, Parkinson’s disease and Alzheimer’s disease. Some of these epigenetic mechanisms include DNA methylation and histone modifications, which have a direct impact on the way that genes are expressed in stem cells and how they drive these cells into a mature lineage. Understanding how the stem cells are behaving and giving rise to mature cells can be used to inform researchers on effective ways to design stem cell-based treatments. In this review article, the way in which the basic understanding of how manipulating this process can be utilized to treat certain neurological diseases will be presented. Different genetic factors and their epigenetic changes during reprogramming of stem cells into induced pluripotent stem cells (iPSCs) have significant potential for enhancing the efficacy of cell replacement therapies.
Collapse
|
34
|
Lardenoije R, van den Hove DL, Vaessen TS, Iatrou A, Meuwissen KP, van Hagen BT, Kenis G, Steinbusch HW, Schmitz C, Rutten BP. Epigenetic modifications in mouse cerebellar Purkinje cells: effects of aging, caloric restriction, and overexpression of superoxide dismutase 1 on 5-methylcytosine and 5-hydroxymethylcytosine. Neurobiol Aging 2015; 36:3079-3089. [DOI: 10.1016/j.neurobiolaging.2015.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 07/28/2015] [Accepted: 08/01/2015] [Indexed: 12/20/2022]
|
35
|
Sherwani SI, Khan HA. Role of 5-hydroxymethylcytosine in neurodegeneration. Gene 2015; 570:17-24. [PMID: 26115768 DOI: 10.1016/j.gene.2015.06.052] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 05/18/2015] [Accepted: 06/18/2015] [Indexed: 02/05/2023]
Abstract
The recent discovery of 5-hydroxymethylcytosine (5hmC), an epigenetic modifier and oxidation product of 5-methylcytosine (5mC), has broadened the scope and understanding of neural development and neurodegenerative diseases. By virtue of their functional groups, 5mC and 5hmC exert opposite effects on gene expression; the former is generally associated with gene silencing whereas the latter is mainly involved in up-regulation of gene expression affecting the cellular processes such as differentiation, development, and aging. Although DNA methylation plays an important role in normal neural development and neuroprotection, an altered pathway due to complex interaction with environmental and genetic factors may cause severe neurodegeneration. The levels of 5hmC in brain increase progressively from birth until death, while in patients with neurodegenerative disorders, the levels are found to be highly compromised. This article discusses the recent developments in the area of hydroxymethylation, with particular emphasis on the role of 5hmC in neurodegenerative diseases including Alzheimer's disease, Parkinson's diseases and Huntington's disease. We have also included recent findings on the role of 5hmC in brain tumors (gliomas). Despite compelling evidence on the involvement of 5hmC in neurodegeneration, it is yet to be established whether this epigenetic molecule is the cause or the effect of the onset and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shariq I Sherwani
- Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
36
|
Ortega-Martínez S. A new perspective on the role of the CREB family of transcription factors in memory consolidation via adult hippocampal neurogenesis. Front Mol Neurosci 2015; 8:46. [PMID: 26379491 PMCID: PMC4549561 DOI: 10.3389/fnmol.2015.00046] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/11/2015] [Indexed: 12/21/2022] Open
Abstract
Adult neurogenesis is the process by which new neurons are generated in the brains of adults. Since its discovery 50 years ago, adult neurogenesis has been widely studied in the mammalian brain and has provided a new perspective on the pathophysiology of many psychiatric and neurodegenerative disorders, some of which affect memory. In this regard, adult hippocampal neurogenesis (AHN), which occurs in the subgranular zone (SGZ) of the dentate gyrus (DG), has been suggested to play a role in the formation and consolidation of new memories. This process involves many transcription factors, of which cyclic AMP (cAMP)-responsive element-binding protein (CREB) is a well-documented one. In the developing brain, CREB regulates crucial cell stages (e.g., proliferation, differentiation, and survival), and in the adult brain, it participates in neuronal plasticity, learning, and memory. In addition, new evidence supports the hypothesis that CREB may also participate in learning and memory through its involvement in AHN. This review examines the CREB family of transcription factors, including the different members and known signaling pathways. It highlights the role of CREB as a modulator of AHN, which could underlie its function in memory consolidation mechanisms.
Collapse
Affiliation(s)
- Sylvia Ortega-Martínez
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| |
Collapse
|
37
|
Gramowski-Voß A, Schwertle HJ, Pielka AM, Schultz L, Steder A, Jügelt K, Axmann J, Pries W. Enhancement of Cortical Network Activity in vitro and Promotion of GABAergic Neurogenesis by Stimulation with an Electromagnetic Field with a 150 MHz Carrier Wave Pulsed with an Alternating 10 and 16 Hz Modulation. Front Neurol 2015; 6:158. [PMID: 26236278 PMCID: PMC4500930 DOI: 10.3389/fneur.2015.00158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/25/2015] [Indexed: 12/21/2022] Open
Abstract
In recent years, various stimuli were identified capable of enhancing neurogenesis, a process which is dysfunctional in the senescent brain and in neurodegenerative and certain neuropsychiatric diseases. Applications of electromagnetic fields to brain tissue have been shown to affect cellular properties and their importance for therapies in medicine is recognized. In this study, differentiating murine cortical networks on multiwell microelectrode arrays were repeatedly exposed to an extremely low-electromagnetic field (ELEMF) with alternating 10 and 16 Hz frequencies piggy backed onto a 150 MHz carrier frequency. The ELEMF exposure stimulated the electrical network activity and intensified the structure of bursts. Further, the exposure to electromagnetic fields within the first 28 days in vitro of the differentiation of the network activity induced also reorganization within the burst structure. This effect was already most pronounced at 14 days in vitro after 10 days of exposure. Overall, the development of cortical activity under these conditions was accelerated. These functional electrophysiological changes were accompanied by morphological ones. The percentage of neurons in the neuron glia co-culture was increased without affecting the total number of cells, indicating an enhancement of neurogenesis. The ELEMF exposure selectively promoted the proliferation of a particular population of neurons, evidenced by the increased proportion of GABAergic neurons. The results support the initial hypothesis that this kind of ELEMF stimulation could be a treatment option for specific indications with promising potential for CNS applications, especially for degenerative diseases, such as Alzheimer’s disease and other dementias.
Collapse
Affiliation(s)
| | | | | | - Luise Schultz
- Division of Electrophysiology, NeuroProof GmbH , Rostock , Germany ; Division of Molecular Biology, NeuroProof GmbH , Rostock , Germany
| | - Anne Steder
- Division of Electrophysiology, NeuroProof GmbH , Rostock , Germany
| | | | - Jürgen Axmann
- Engineering Office for Bioresonance and Environmental Technology , Werder/Havel , Germany
| | | |
Collapse
|
38
|
Du N, Xu D, Hou X, Song X, Liu C, Chen Y, Wang Y, Li X. Inverse Association Between Serum Uric Acid Levels and Alzheimer's Disease Risk. Mol Neurobiol 2015; 53:2594-9. [PMID: 26084440 DOI: 10.1007/s12035-015-9271-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
Abstract
The association between Alzheimer's disease and uric acid levels had gained great interest in recent years, but there was still lack of definite evidence. A systematic review and meta-analysis of relevant studies was performed to comprehensively estimate the association. Relevant studies published before October 26, 2014, were searched in PubMed, Embase, and China Biology Medicine (CBM) databases. Study-specific data were combined using random-effects or fixed-effects models of meta-analysis according to between-study heterogeneity. Twenty-four studies (21 case-control and 3 cohort studies) were finally included into the meta-analysis. Those 21 case-control studies included a total of 1128 cases of Alzheimer's disease and 2498 controls without Alzheimer's disease. Those 3 cohort studies included a total of 7327 participants. Meta-analysis showed that patients with Alzheimer's disease had lower levels of uric acid than healthy controls (weighted mean difference (WMD) = -0.77 mg/dl, 95% CI -2.28 to -0.36, P = 0.0002). High serum uric acid levels were significantly associated with decreased risk of Alzheimer's disease (risk ratio (RR) = 0.66, 95% CI 0.52-0.85, P = 0.001). There was low risk of publication bias in the meta-analysis. There is an inverse association between serum uric acid levels and Alzheimer's disease. High serum uric acid level is a protective factor of Alzheimer's disease.
Collapse
Affiliation(s)
- Na Du
- Department of Endocrinology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Donghua Xu
- Department of Rheumatology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| | - Xu Hou
- Department of Endocrinology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Xuejia Song
- Department of Endocrinology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Cancan Liu
- Department of Endocrinology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Ying Chen
- Department of Endocrinology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yangang Wang
- Department of Endocrinology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
| | - Xin Li
- Department of Physiology, Functional Laboratory of Weifang Medical University, Weifang, 261031, China.
| |
Collapse
|
39
|
Chu W, Yuan J, Huang L, Xiang X, Zhu H, Chen F, Chen Y, Lin J, Feng H. Valproic Acid Arrests Proliferation but Promotes Neuronal Differentiation of Adult Spinal NSPCs from SCI Rats. Neurochem Res 2015; 40:1472-86. [PMID: 26023063 DOI: 10.1007/s11064-015-1618-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 04/02/2015] [Accepted: 05/18/2015] [Indexed: 11/29/2022]
Abstract
Although the adult spinal cord contains a population of multipotent neural stem/precursor cells (NSPCs) exhibiting the potential to replace neurons, endogenous neurogenesis is very limited after spinal cord injury (SCI) because the activated NSPCs primarily differentiate into astrocytes rather than neurons. Valproic acid (VPA), a histone deacetylase inhibitor, exerts multiple pharmacological effects including fate regulation of stem cells. In this study, we cultured adult spinal NSPCs from chronic compressive SCI rats and treated with VPA. In spite of inhibiting the proliferation and arresting in the G0/G1 phase of NSPCs, VPA markedly promoted neuronal differentiation (β-tubulin III(+) cells) as well as decreased astrocytic differentiation (GFAP(+) cells). Cell cycle regulator p21(Cip/WAF1) and proneural genes Ngn2 and NeuroD1 were increased in the two processes respectively. In vivo, to minimize the possible inhibitory effects of VPA to the proliferation of NSPCs as well as avoid other neuroprotections of VPA in acute phase of SCI, we carried out a delayed intraperitoneal injection of VPA (150 mg/kg/12 h) to SCI rats from day 15 to day 22 after injury. Both of the newborn neuron marker doublecortin and the mature neuron marker neuron-specific nuclear protein were significantly enhanced after VPA treatment in the epicenter and adjacent segments of the injured spinal cord. Although the impaired corticospinal tracks had not significantly improved, Basso-Beattie-Bresnahan scores in VPA treatment group were better than control. Our study provide the first evidence that administration of VPA enhances the neurogenic potential of NSPCs after SCI and reveal the therapeutic value of delayed treatment of VPA to SCI.
Collapse
Affiliation(s)
- Weihua Chu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 29, Gaotanyan Street, Shapingba District, Chongqing, 400038, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Venkatesan R, Ji E, Kim SY. Phytochemicals that regulate neurodegenerative disease by targeting neurotrophins: a comprehensive review. BIOMED RESEARCH INTERNATIONAL 2015; 2015:814068. [PMID: 26075266 PMCID: PMC4446472 DOI: 10.1155/2015/814068] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/17/2015] [Accepted: 04/24/2015] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), characterized by progressive dementia and deterioration of cognitive function, is an unsolved social and medical problem. Age, nutrition, and toxins are the most common causes of AD. However, currently no credible treatment is available for AD. Traditional herbs and phytochemicals may delay its onset and slow its progression and also allow recovery by targeting multiple pathological causes by antioxidative, anti-inflammatory, and antiamyloidogenic properties. They also regulate mitochondrial stress, apoptotic factors, free radical scavenging system, and neurotrophic factors. Neurotrophins such as BDNF, NGF, NT3, and NT4/5 play a vital role in neuronal and nonneuronal responses to AD. Neurotrophins depletion accelerates the progression of AD and therefore, replacing such neurotrophins may be a potential treatment for neurodegenerative disease. Here, we review the phytochemicals that mediate the signaling pathways involved in neuroprotection specifically neurotrophin-mediated activation of Trk receptors and members of p75(NTR) superfamily. We focus on representative phenolic derivatives, iridoid glycosides, terpenoids, alkaloids, and steroidal saponins as regulators of neurotrophin-mediated neuroprotection. Although these phytochemicals have attracted attention owing to their in vitro neurotrophin potentiating activity, their in vivo and clinical efficacy trials has yet to be established. Therefore, further research is necessary to prove the neuroprotective effects in preclinical models and in humans.
Collapse
Affiliation(s)
- Ramu Venkatesan
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 406-799, Republic of Korea
| | - Eunhee Ji
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 406-799, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 406-799, Republic of Korea
- Gachon Medical Research Institute, Gil Medical Center, Inchon 405-760, Republic of Korea
- Gachon Institute of Pharmaceutical Science, Gachon University, No. 191 Hambakmoe-ro, Yeonsu-gu, Incheon 406-799, Republic of Korea
| |
Collapse
|
41
|
Quantification of histone deacetylase isoforms in human frontal cortex, human retina, and mouse brain. PLoS One 2015; 10:e0126592. [PMID: 25962138 PMCID: PMC4427357 DOI: 10.1371/journal.pone.0126592] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/06/2015] [Indexed: 12/30/2022] Open
Abstract
Histone deacetylase (HDAC) inhibition has promise as a therapy for Alzheimer’s disease (AD) and other neurodegenerative diseases. Currently, therapeutic HDAC inhibitors target many HDAC isoforms, a particularly detrimental approach when HDAC isoforms are known to have different and specialized functions. We have developed a multiple reaction monitoring (MRM) mass spectrometry assay using stable isotope-labeled QconCATs as internal standards to quantify HDAC isoforms. We further determined a quantitative pattern of specific HDACs expressed in various human and mouse neural tissues. In human AD frontal cortex, HDAC1,2 decreased 32%, HDAC5 increased 47%, and HDAC6 increased 31% in comparison to age-matched controls. Human neural retina concentrations of HDAC1, 2, HDAC5, HDAC6, and HDAC7 decreased in age-related macular degeneration (AMD)-affected donors and exhibited a greater decrease in AD-affected donors in comparison to age-matched control neural retinas. Additionally, HDAC concentrations were measured in whole hemisphere of brain of 5XFAD mice, a model of β-amyloid deposition, to assess similarity to AD in human frontal cortex. HDAC profiles of human frontal cortex and mouse hemisphere had noticeable differences and relatively high concentrations of HDAC3 and HDAC4 in mice, which were undetectable in humans. Our method for quantification of HDAC isoforms is a practical and efficient technique to quantify isoforms in various tissues and diseases. Changes in HDAC concentrations reported herein contribute to the understanding of the pathology of neurodegeneration.
Collapse
|
42
|
Li XY, Bao XJ, Wang RZ. Potential of neural stem cell-based therapies for Alzheimer's disease. J Neurosci Res 2015; 93:1313-24. [PMID: 25601591 DOI: 10.1002/jnr.23555] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/23/2014] [Accepted: 12/15/2014] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD), known to be a leading cause of dementia that causes heavy social and financial burdens worldwide, is characterized by progressive loss of neurons and synaptic connectivity after depositions of amyloid-β (Aβ) protein. Current therapies for AD patients can only alleviate symptoms but cannot deter the neural degeneration, thus providing no long-term recovery. Neural stem cells (NSCs), capable of self-renewal and of differentiation into functional neurons and glia, have been shown to repair damaged networks and reverse memory and learning deficits in animal studies, providing new hope for curing AD patients by cell transplantation. Under AD pathology, the microenvironment also undergoes great alterations that affect the propagation of NSCs and subsequent therapeutic efficiency, calling for measures to improve the hostile environment for cell transplantation. This article reviews the therapeutic potential of both endogenous and exogenous NSCs in the treatment of AD and the challenges to application of stem cells in AD treatment, particularly those from the microenvironmental alterations, in the hope of providing more information for future research in exploiting stem cell-based therapies for AD. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xue-Yuan Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Xin-Jie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Ren-Zhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|