1
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
2
|
Bai Y, Wang Z, Yu L, Dong K, Cheng L, Zhu R. The enhanced generation of motor neurons from mESCs by MgAl layered double hydroxide nanoparticles. Biomed Mater 2023; 18. [PMID: 36898160 DOI: 10.1088/1748-605x/acc375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/10/2023] [Indexed: 03/12/2023]
Abstract
The committed differentiation of stem cells into neurons is a promising therapeutic strategy for neurological diseases. Predifferentiation of transplanted stem cells into neural precursors could enhance their utilization and control the direction of differentiation. Embryonic stem cells with totipotency can differentiate into specific nerve cells under appropriate external induction conditions. Layered double hydroxide (LDH) nanoparticles have been proven to regulate the pluripotency of mouse ESCs (mESCs), and LDH could be used as carrier in neural stem cells for nerve regeneration. Hence, we sought to study the effects of LDH without loaded factors on mESCs neurogenesis in this work. A series of characteristics analyses indicated the successful construction of LDH nanoparticles. LDH nanoparticles that may adhere to the cell membranes had insignificant effect on cell proliferation and apoptosis. The enhanced differentiation of mESCs into motor neurons by LDH was systematically validated by immunofluorescent staining, quantitative real-time PCR analysis and western blot analysis. In addition, transcriptome sequencing analysis and mechanism verification elucidated the significant regulatory roles of focal adhesion signaling pathway in the enhanced mESCs neurogenesis by LDH. Taken together, the functional validation of inorganic LDH nanoparticles promoting motor neurons differentiation provide a novel strategy and therapeutic prospect for the clinical transition of neural regeneration.
Collapse
Affiliation(s)
- Yuxin Bai
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai 200065, People's Republic of China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, People's Republic of China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai 200065, People's Republic of China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, People's Republic of China
| | - Liqun Yu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai 200065, People's Republic of China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, People's Republic of China
| | - Kun Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai 200065, People's Republic of China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, People's Republic of China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai 200065, People's Republic of China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, People's Republic of China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai 200065, People's Republic of China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, People's Republic of China
| |
Collapse
|
3
|
Mokhtar DM, Sayed RKA, Zaccone G, Albano M, Hussein MT. Ependymal and Neural Stem Cells of Adult Molly Fish ( Poecilia sphenops, Valenciennes, 1846) Brain: Histomorphometry, Immunohistochemical, and Ultrastructural Studies. Cells 2022; 11:2659. [PMID: 36078068 PMCID: PMC9455025 DOI: 10.3390/cells11172659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/11/2022] [Accepted: 08/24/2022] [Indexed: 12/18/2022] Open
Abstract
This study was conducted on 16 adult specimens of molly fish (Poecilia sphenops) to investigate ependymal cells (ECs) and their role in neurogenesis using ultrastructural examination and immunohistochemistry. The ECs lined the ventral and lateral surfaces of the optic ventricle and their processes extended through the tectal laminae and ended at the surface of the tectum as a subpial end-foot. Two cell types of ECs were identified: cuboidal non-ciliated (5.68 ± 0.84/100 μm2) and columnar ciliated (EC3.22 ± 0.71/100 μm2). Immunohistochemical analysis revealed two types of GFAP immunoreactive cells: ECs and astrocytes. The ECs showed the expression of IL-1β, APG5, and Nfr2. Moreover, ECs showed immunostaining for myostatin, S100, and SOX9 in their cytoplasmic processes. The proliferative activity of the neighboring stem cells was also distinct. The most interesting finding in this study was the glia-neuron interaction, where the processes of ECs met the progenitor neuronal cells in the ependymal area of the ventricular wall. These cells showed bundles of intermediate filaments in their processes and basal poles and were connected by desmosomes, followed by gap junctions. Many membrane-bounded vesicles could be demonstrated on the surface of the ciliated ECs that contained neurosecretion. The abluminal and lateral cell surfaces of ECs showed pinocytotic activities with many coated vesicles, while their apical cytoplasm contained centrioles. The occurrence of stem cells in close position to the ECs, and the presence of bundles of generating axons in direct contact with these stem cells indicate the role of ECs in neurogenesis. The TEM results revealed the presence of neural stem cells in a close position to the ECs, in addition to the presence of bundles of generating axons in direct contact with these stem cells. The present study indicates the role of ECs in neurogenesis.
Collapse
Affiliation(s)
- Doaa M. Mokhtar
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assuit University, Assiut 71526, Egypt
| | - Ramy K. A. Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| | - Giacomo Zaccone
- Department of Veterinary Sciences, Polo Universitario dell’Annunziata, University of Messina, 98168 Messina, Italy
| | - Marco Albano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Manal T. Hussein
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assuit University, Assiut 71526, Egypt
| |
Collapse
|
4
|
Higo N. Motor Cortex Plasticity During Functional Recovery Following Brain Damage. JOURNAL OF ROBOTICS AND MECHATRONICS 2022. [DOI: 10.20965/jrm.2022.p0700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although brain damage causes functional impairment, it is often followed by partial or total recovery of function. Recovery is believed to occur primarily because of brain plasticity. Both human and animal studies have significantly contributed to uncovering the neuronal basis of plasticity. Recent advances in brain imaging technology have enabled the investigation of plastic changes in living human brains. In addition, animal experiments have revealed detailed changes at the neural and genetic levels. In this review, plasticity in motor-related areas of the cerebral cortex, which is one of the most well-studied areas of the neocortex in terms of plasticity, is reviewed. In addition, the potential of technological interventions to enhance plasticity and promote functional recovery following brain damage is discussed. Novel neurorehabilitation technologies are expected to be established based on the emerging research on plasticity from the last several decades.
Collapse
|
5
|
Wang J, Zhang Z, Fu S, Li X, Li X, Wang S, Yuan L. Overexpression of EphB4 promotes neurogenesis, but inhibits neuroinflammation in mice with acute ischemic stroke. Mol Med Rep 2021; 24:756. [PMID: 34476505 PMCID: PMC8436223 DOI: 10.3892/mmr.2021.12396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/10/2021] [Indexed: 11/25/2022] Open
Abstract
Ischemic stroke is one of the most common diseases that has a high rate of mortality, and has become a burden to the healthcare system. Previous research has shown that EPH receptor B4 (EphB4) promotes neural stem cell proliferation and differentiation in vitro. However, little is known regarding its role in the neurogenesis of ischemic stroke in vivo. Thus, the present study aimed to verify whether EphB4 was a key regulator of neurogenesis in ischemic stroke in vivo. Cerebral ischemia was induced in C57BL/6J mice via middle cerebral artery occlusion (MCAO), followed by reperfusion. Immunofluorescence staining was performed to evaluate the effect of EphB4 on the neurogenesis in cerebral cortex. The levels of inflammatory cytokines were determined using an ELISA kit. The expression levels of ABL proto-oncogene 1, non-receptor tyrosine kinase (ABL1)/Cyclin D1 signaling pathway-related proteins were detected via western blotting. The current findings indicated that EphB4 expression was significantly increased in the cerebral cortex of MCAO model mice in comparison with sham-operated mice. Moreover, EphB4 appeared to be expressed in neural stem cells (Nestin+), and persisted as these cells became neuronal progenitors (Sox2+), neuroblasts [doublecortin (DCX)+], and eventually mature neurons [neuronal nuclei (NeuN)+]. Overexpression of EphB4 elevated the number of proliferating (bromodeoxyuridine+, Ki67+) and differentiated cells (Nestin+, Sox2+, DCX+ and NeuN+), indicating the promoting effect of EphB4 on the neurogenesis of ischemic stroke. Furthermore, EphB4 overexpression alleviated the inflammation injury in MCAO model mice. The expression levels of proteins-related to the ABL1/Cyclin D1 signaling pathway were significantly increased by the overexpression of EphB4, which suggested that restoration of EphB4 promoted the activation of the ABL1/Cyclin D1 signaling pathway. In conclusion, this study contributes to the current understanding of the mechanisms of EphB4 in exerting neurorestorative effects and may recommend a potential new strategy for ischemic stroke treatment.
Collapse
Affiliation(s)
- Jin Wang
- Department of Neurology, Inner Mongolia Baogang Hospital, Baotou, Inner Mongolia 014010, P.R. China
| | - Zun Zhang
- Department of Orthopedics, Inner Mongolia Baogang Hospital, Baotou, Inner Mongolia 014010, P.R. China
| | - Shaojing Fu
- Department of Neurology, Inner Mongolia Baogang Hospital, Baotou, Inner Mongolia 014010, P.R. China
| | - Xiaojie Li
- Department of Neurology, Inner Mongolia Baogang Hospital, Baotou, Inner Mongolia 014010, P.R. China
| | - Xinhui Li
- Department of Neurology, First Affiliated Hospital of Baotou Medical College, Baotou, Inner Mongolia 014016, P.R. China
| | - Shaobin Wang
- Department of Neurology, Inner Mongolia Baogang Hospital, Baotou, Inner Mongolia 014010, P.R. China
| | - Lihe Yuan
- Department of Neurology, Inner Mongolia Baogang Hospital, Baotou, Inner Mongolia 014010, P.R. China
| |
Collapse
|
6
|
Saini A, Singh J, Kumar S. Optically superior fluorescent probes for selective imaging of cells, tumors, and reactive chemical species. Org Biomol Chem 2021; 19:5208-5236. [PMID: 34037048 DOI: 10.1039/d1ob00509j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fluorescent chemical probes have become powerful tools to study biological events in living cells. They provide a great opportunity to quantitatively and qualitatively analyze the physiological and biochemical properties of living cells in real time. The ability of researchers to manipulate these probes for a desired specific purpose has turned many heads in the scientific community. Despite a slow start, fluorescent probe research has seen exponential growth over the last decade in the world. This change required some adventurous and creative scientists from different fields-like biology, medicine, and chemistry-to come together to facilitate the constant expansion of this field. This review article introduces some fundamental concepts related to fluorescent probe designing and development. It also summarizes various fluorescent probes with superior optical properties used in fields like cell biology, cellular imaging, medical research, and cancer diagnosis. It is hoped that this article will encourage more young and creative scientists to contribute their talents to this field.
Collapse
Affiliation(s)
- Abhishek Saini
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India.
| | - Jyoti Singh
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India.
| | - Sonu Kumar
- Department of Chemistry, Hansraj College, University of Delhi, Delhi-110007, India.
| |
Collapse
|
7
|
Butruille L, Vancamp P, Demeneix BA, Remaud S. Thyroid hormone regulation of adult neural stem cell fate: A comparative analysis between rodents and primates. VITAMINS AND HORMONES 2021; 116:133-192. [PMID: 33752817 DOI: 10.1016/bs.vh.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thyroid hormone (TH) signaling, a highly conserved pathway across vertebrates, is crucial for brain development and function throughout life. In the adult mammalian brain, including that of humans, multipotent neural stem cells (NSCs) proliferate and generate neuronal and glial progenitors. The role of TH has been intensively investigated in the two main neurogenic niches of the adult mouse brain, the subventricular and the subgranular zone. A key finding is that T3, the biologically active form of THs, promotes NSC commitment toward a neuronal fate. In this review, we first discuss the roles of THs in the regulation of adult rodent neurogenesis, as well as how it relates to functional behavior, notably olfaction and cognition. Most research uncovering these roles of TH in adult neurogenesis was conducted in rodents, whose genetic background, brain structure and rate of neurogenesis are considerably different from that of humans. To bridge the phylogenetic gap, we also explore the similarities and divergences of TH-dependent adult neurogenesis in non-human primate models. Lastly, we examine how photoperiodic length changes TH homeostasis, and how that might affect adult neurogenesis in seasonal species to increase fitness. Several aspects by which TH acts on adult NSCs seem to be conserved among mammals, while we only start to uncover the molecular pathways, as well as how other in- and extrinsic factors are intertwined. A multispecies approach delivering more insights in the matter will pave the way for novel NSC-based therapies to combat neurological disorders.
Collapse
Affiliation(s)
- Lucile Butruille
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Pieter Vancamp
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France.
| |
Collapse
|
8
|
Jinnou H. Regeneration using endogenous neural stem cells following neonatal brain injury. Pediatr Int 2021; 63:13-21. [PMID: 32609915 DOI: 10.1111/ped.14368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 05/29/2020] [Accepted: 06/25/2020] [Indexed: 01/25/2023]
Abstract
Despite recent advancements in perinatal care, the incidence of neonatal brain injury has not decreased. No therapies are currently available to repair injured brain tissues. In the postnatal brain, neural stem cells reside in the ventricular-subventricular zone (V-SVZ) and continuously generate new immature neurons (neuroblasts). After brain injury in rodents, V-SVZ-derived neuroblasts migrate toward the injured area using blood vessels as a scaffold. Notably, the neonatal V-SVZ has a remarkable neurogenic capacity. Furthermore, compared with the adult brain, after neonatal brain injury, larger numbers of neuroblasts migrate toward the lesion, raising the possibility that the V-SVZ could be a source for endogenous neuronal regeneration after neonatal brain injury. We recently demonstrated that efficient migration of V-SVZ-derived neuroblasts toward a lesion is supported by neonatal radial glia via neural cadherin (N-cadherin)-mediated neuron-fiber contact, which promotes RhoA activity. Moreover, providing blood vessel- and radial glia-mimetic scaffolds for migrating neuroblasts promotes neuronal migration and improves functional gait behaviors after neonatal brain injury. In the V-SVZ, oligodendrocyte progenitor cells (OPCs) are also generated and migrate toward the surrounding white matter, where they differentiate and form myelin. After white matter injury in rodents, the production and subsequent migration of V-SVZ-derived OPCs are enhanced. In the neonatal period, administration of growth factors at a specific time promotes oligodendrocyte regeneration and functional recovery after brain injury. These findings suggest that activating the high regenerative capacity that is specific to the neonatal period could lead to the development of new therapeutic strategies for neonatal brain injury.
Collapse
Affiliation(s)
- Hideo Jinnou
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
9
|
Flanagan A, Bechtold DA, Pot GK, Johnston JD. Chrono-nutrition: From molecular and neuronal mechanisms to human epidemiology and timed feeding patterns. J Neurochem 2020; 157:53-72. [PMID: 33222161 DOI: 10.1111/jnc.15246] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
The circadian timing system governs daily biological rhythms, synchronising physiology and behaviour to the temporal world. External time cues, including the light-dark cycle and timing of food intake, provide daily signals for entrainment of the central, master circadian clock in the hypothalamic suprachiasmatic nuclei (SCN), and of metabolic rhythms in peripheral tissues, respectively. Chrono-nutrition is an emerging field building on the relationship between temporal eating patterns, circadian rhythms, and metabolic health. Evidence from both animal and human research demonstrates adverse metabolic consequences of circadian disruption. Conversely, a growing body of evidence indicates that aligning food intake to periods of the day when circadian rhythms in metabolic processes are optimised for nutrition may be effective for improving metabolic health. Circadian rhythms in glucose and lipid homeostasis, insulin responsiveness and sensitivity, energy expenditure, and postprandial metabolism, may favour eating patterns characterised by earlier temporal distribution of energy. This review details the molecular basis for metabolic clocks, the regulation of feeding behaviour, and the evidence for meal timing as an entraining signal for the circadian system in animal models. The epidemiology of temporal eating patterns in humans is examined, together with evidence from human intervention studies investigating the metabolic effects of morning compared to evening energy intake, and emerging chrono-nutrition interventions such as time-restricted feeding. Chrono-nutrition may have therapeutic application for individuals with and at-risk of metabolic disease and convey health benefits within the general population.
Collapse
Affiliation(s)
- Alan Flanagan
- Section of Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,Section of Metabolic Medicine, Food and Macronutrients, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - David A Bechtold
- Division of Diabetes, Endocrinology & Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Gerda K Pot
- Department of Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Nutrition and Health Department, Louis Bolk Instituut, Bunnik, the Netherlands
| | - Jonathan D Johnston
- Section of Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
10
|
Synthesis, structural investigations, DFT studies, and neurotrophic activity of zinc complex with a multidentate ligand. MONATSHEFTE FUR CHEMIE 2020. [DOI: 10.1007/s00706-020-02696-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Deboux C, Spigoni G, Caillava C, Garcia-Diaz B, Ypsilanti A, Sarrazin N, Bachelin C, Chédotal A, Baron-Van Evercooren A. Slit1 Protein Regulates SVZ-Derived Precursor Mobilization in the Adult Demyelinated CNS. Front Cell Neurosci 2020; 14:168. [PMID: 32670024 PMCID: PMC7332780 DOI: 10.3389/fncel.2020.00168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023] Open
Abstract
Slit1 is a secreted axon guidance molecule, also involved in adult neurogenesis. In physiological conditions, Slit1 loss promotes ectopic dispersal of SVZ-derived neural precursors (SVZ-NPCs) into periventricular structures such as the corpus callosum. Demyelination of the corpus callosum triggers SVZ-NPC migration to ectopic locations and their recruitment by the lesion, suggesting a possible role for Slit1 in SVZ-NPCs ectopic dispersal regulation in pathological conditions. Here, we have investigated the function of Slit1 protein in the recruitment of SVZ-NPCs after CNS demyelination. We find that the dynamics of oligodendrogenesis and temporal profile of developmental myelination in Slit1–/– mice are similar to Slit1+/− controls. SVZ micro-dissection and RT-PCR from wild-type mice, show that Slits and Robos are physiologically regulated at the transcriptional level in response to corpus callosum demyelination suggesting their role in the process of SVZ-NPC ectopic migration in demyelinating conditions. Moreover, we find that the number of SVZ-NPCs recruited by the lesion increases in Sli1–/– mice compared to Slit1+/− mice, leading to higher numbers of Olig2+ cells within the lesion. Time-lapse video-microscopy of immuno-purified NPCs shows that Slit1-deficient cells migrate faster and make more frequent directional changes than control NPCs, supporting a cell-autonomous mechanism of action of Slit1 in NPC migration. In conclusion, while Slit1 does not affect the normal developmental process of oligodendrogenesis and myelination, it regulates adult SVZ-NPC ectopic migration in response to demyelination, and consequently oligodendrocyte renewal within the lesion.
Collapse
Affiliation(s)
- C Deboux
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - G Spigoni
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Caillava
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - B Garcia-Diaz
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Ypsilanti
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - N Sarrazin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Bachelin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Chédotal
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - A Baron-Van Evercooren
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| |
Collapse
|
12
|
Gothié J, Vancamp P, Demeneix B, Remaud S. Thyroid hormone regulation of neural stem cell fate: From development to ageing. Acta Physiol (Oxf) 2020; 228:e13316. [PMID: 31121082 PMCID: PMC9286394 DOI: 10.1111/apha.13316] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 12/13/2022]
Abstract
In the vertebrate brain, neural stem cells (NSCs) generate both neuronal and glial cells throughout life. However, their neuro‐ and gliogenic capacity changes as a function of the developmental context. Despite the growing body of evidence on the variety of intrinsic and extrinsic factors regulating NSC physiology, their precise cellular and molecular actions are not fully determined. Our review focuses on thyroid hormone (TH), a vital component for both development and adult brain function that regulates NSC biology at all stages. First, we review comparative data to analyse how TH modulates neuro‐ and gliogenesis during vertebrate brain development. Second, as the mammalian brain is the most studied, we highlight the molecular mechanisms underlying TH action in this context. Lastly, we explore how the interplay between TH signalling and cell metabolism governs both neurodevelopmental and adult neurogenesis. We conclude that, together, TH and cellular metabolism regulate optimal brain formation, maturation and function from early foetal life to adult in vertebrate species.
Collapse
Affiliation(s)
- Jean‐David Gothié
- Department of Neurology & Neurosurgery Montreal Neurological Institute & Hospital, McGill University Montreal Quebec Canada
| | - Pieter Vancamp
- CNRS UMR 7221 Muséum National d’Histoire Naturelle Paris France
| | | | - Sylvie Remaud
- CNRS UMR 7221 Muséum National d’Histoire Naturelle Paris France
| |
Collapse
|
13
|
Dief AE, Hassan PS, Hartmut O, Jirikowski GF. Neuronal and glial regeneration after focal cerebral ischemia in rat, an immunohistochemical and electron microscopical study. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Abeer E. Dief
- Department of Physiology, University of Alexandria, Egypt
| | | | | | | |
Collapse
|
14
|
Structural and biological evaluation of a platinum complex as a potential anti-neurodegenerative agent. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
15
|
Silveira GDP, Ishimura ME, Teixeira D, Galindo LT, Sardinha AA, Porcionatto M, Longo-Maugéri IM. Improvement of Mesenchymal Stem Cell Immunomodulatory Properties by Heat-Killed Propionibacterium acnes via TLR2. Front Mol Neurosci 2019; 11:489. [PMID: 30687005 PMCID: PMC6336115 DOI: 10.3389/fnmol.2018.00489] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are an essential tool for regenerative medicine, which aims to develop new technologies to improve their effects to obtain useful transplantation results. MSC immunomodulatory role has been just demonstrated; however, how they react when they are stimulated by an adjuvant is poorly understood. Our group showed the adjuvant effect of killed Propionibacterium acnes (P. acnes) on hematopoietic stem cells. As these cells share the same MSCs bone marrow (BM) site and interact with each other, here we evaluated the P. acnes and its soluble polysaccharide (PS) effect on MSCs and their immunomodulatory role in a murine model of traumatic brain injury (TBI). The bacteria increased the absolute number of MSCs, including MSC subpopulations, and maintained MSC plasticity. P. acnes and PS enhanced MSC proliferation and improved their immunomodulatory effect. P. acnes-MSC and PS-MSC transplantation increased anti-inflammatory cytokine expression and diminished pro-inflammatory cytokine expression after injury. This effect seemed to be mediated via TLR2 since P. acnes-KOTLR2-MSC transplantation decreased TGF-β and IL-10 expression. Increasing in neural stem cells and neuroblasts after PS-MSC transplantation was also observed. The adjuvant effect of P. acnes is an alternative means of expanding MSCs and important to identify their subpopulations to know better their role under exogenous stimuli including inflammation resolution in an experimental model.
Collapse
Affiliation(s)
- Gabriela da Paz Silveira
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Mayari Eika Ishimura
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela Teixeira
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Layla Tesla Galindo
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Agnes Araujo Sardinha
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Marimelia Porcionatto
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ieda Maria Longo-Maugéri
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
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: 6.8] [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
|
17
|
Gothié JD, Demeneix B, Remaud S. Comparative approaches to understanding thyroid hormone regulation of neurogenesis. Mol Cell Endocrinol 2017; 459:104-115. [PMID: 28545819 DOI: 10.1016/j.mce.2017.05.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/11/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
Thyroid hormone (TH) signalling, an evolutionary conserved pathway, is crucial for brain function and cognition throughout life, from early development to ageing. In humans, TH deficiency during pregnancy alters offspring brain development, increasing the risk of cognitive disorders. How TH regulates neurogenesis and subsequent behaviour and cognitive functions remains a major research challenge. Cellular and molecular mechanisms underlying TH signalling on proliferation, survival, determination, migration, differentiation and maturation have been studied in mammalian animal models for over a century. However, recent data show that THs also influence embryonic and adult neurogenesis throughout vertebrates (from mammals to teleosts). These latest observations raise the question of how TH availability is controlled during neurogenesis and particularly in specific neural stem cell populations. This review deals with the role of TH in regulating neurogenesis in the developing and the adult brain across different vertebrate species. Such evo-devo approaches can shed new light on (i) the evolution of the nervous system and (ii) the evolutionary control of neurogenesis by TH across animal phyla. We also discuss the role of thyroid disruptors on brain development in an evolutionary context.
Collapse
Affiliation(s)
- Jean-David Gothié
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France
| | - Barbara Demeneix
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France.
| | - Sylvie Remaud
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France.
| |
Collapse
|
18
|
Alonso MI, Lamus F, Carnicero E, Moro JA, de la Mano A, Fernández JMF, Desmond ME, Gato A. Embryonic Cerebrospinal Fluid Increases Neurogenic Activity in the Brain Ventricular-Subventricular Zone of Adult Mice. Front Neuroanat 2017; 11:124. [PMID: 29311854 PMCID: PMC5742215 DOI: 10.3389/fnana.2017.00124] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022] Open
Abstract
Neurogenesis is a very intensive process during early embryonic brain development, becoming dramatically restricted in the adult brain in terms of extension and intensity. We have previously demonstrated the key role of embryonic cerebrospinal fluid (CSF) in developing brain neurogenic activity. We also showed that cultured adult brain neural stem cells (NSCs) remain competent when responding to the neurogenic influence of embryonic CSF. However, adult CSF loses its neurogenic inductive properties. Here, by means of an organotypic culture of adult mouse brain sections, we show that local administration of embryonic CSF in the subventricular zone (SVZ) niche is able to trigger a neurogenic program in NSCs. This leads to a significant increase in the number of non-differentiated NSCs, and also in the number of new neurons which show normal migration, differentiation and maturation. These new data reveal that embryonic CSF activates adult brain NSCs, supporting the previous idea that it contains key instructive components which could be useful in adult brain neuroregenerative strategies.
Collapse
Affiliation(s)
- Maria I Alonso
- Departamento de Anatomía y Radiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.,Laboratorio de Desarrollo y Teratología del Sistema Nervioso, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Valladolid, Valladolid, Spain
| | - Francisco Lamus
- Departamento de Anatomía y Radiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Estela Carnicero
- Departamento de Anatomía y Radiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.,Laboratorio de Desarrollo y Teratología del Sistema Nervioso, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Valladolid, Valladolid, Spain
| | - Jose A Moro
- Departamento de Anatomía y Radiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.,Laboratorio de Desarrollo y Teratología del Sistema Nervioso, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Valladolid, Valladolid, Spain
| | - Anibal de la Mano
- Departamento de Anatomía y Radiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.,Laboratorio de Desarrollo y Teratología del Sistema Nervioso, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Valladolid, Valladolid, Spain
| | - Jose M F Fernández
- Departamento de Biología Celular, Histología y Farmacología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Mary E Desmond
- Department of Biology, Villanova University, Villanova, PA, United States
| | - Angel Gato
- Departamento de Anatomía y Radiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.,Laboratorio de Desarrollo y Teratología del Sistema Nervioso, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
19
|
Gómez C, Jimeno D, Fernández-Medarde A, García-Navas R, Calzada N, Santos E. Ras-GRF2 regulates nestin-positive stem cell density and onset of differentiation during adult neurogenesis in the mouse dentate gyrus. Mol Cell Neurosci 2017; 85:127-147. [PMID: 28966131 DOI: 10.1016/j.mcn.2017.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/11/2017] [Accepted: 09/17/2017] [Indexed: 12/12/2022] Open
Abstract
Various parameters of neurogenesis were analyzed in parallel in the two neurogenic areas (the Dentate Gyrus[DG] and the Subventricular Zone[SVZ]/Rostral Migratory Stream[RMS]/Main Olfactory Bulb[MOB] neurogenic system) of adult WT and KO mouse strains for the Ras-GRF1/2 genes (Ras-GRF1-KO, Ras-GRF2-KO, Ras-GRF1/2-DKO). Significantly reduced numbers of doublecortin[DCX]-positive cells were specifically observed in the DG, but not the SVZ/RMS/MOB neurogenic region, of Ras-GRF2-KO and Ras-GRF1/2-DKO mice indicating that this novel Ras-GRF2-dependent phenotype is spatially restricted to a specific neurogenic area. Consistent with a role of CREB as mediator of Ras-GRF2 function in neurogenesis, the density of p-CREB-positive cells was also specifically reduced in all neurogenic regions of Ras-GRF2-KO and DKO mice. Similar levels of early neurogenic proliferation markers (Ki67, BrdU) were observed in all different Ras-GRF genotypes analyzed but significantly elevated levels of nestin-immunolabel, particularly of undifferentiated, highly ramified, A-type nestin-positive neurons were specifically detected in the DG but not the SVZ/RMS/MOB of Ras-GRF2-KO and DKO mice. Together with assays of other neurogenic markers (GFAP, Sox2, Tuj1, NeuN), these observations suggest that the deficit of DCX/p-CREB-positive cells in the DG of Ras-GRF2-depleted mice does not involve impaired neuronal proliferation but rather delayed transition from the stem cell stage to the differentiation stages of the neurogenic process. This model is also supported by functional analyses of DG-derived neurosphere cultures and transcriptional characterization of the neurogenic areas of mice of all relevant Ras-GRF genotypes suggesting that the neurogenic role of Ras-GRF2 is exerted in a cell-autonomous manner through a specific transcriptional program.
Collapse
Affiliation(s)
- Carmela Gómez
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC- Universidad de Salamanca) and CIBERONC, 37007 Salamanca, Spain
| | - David Jimeno
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC- Universidad de Salamanca) and CIBERONC, 37007 Salamanca, Spain
| | - Alberto Fernández-Medarde
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC- Universidad de Salamanca) and CIBERONC, 37007 Salamanca, Spain
| | - Rósula García-Navas
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC- Universidad de Salamanca) and CIBERONC, 37007 Salamanca, Spain
| | - Nuria Calzada
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC- Universidad de Salamanca) and CIBERONC, 37007 Salamanca, Spain
| | - Eugenio Santos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC- Universidad de Salamanca) and CIBERONC, 37007 Salamanca, Spain.
| |
Collapse
|
20
|
Pushchina EV, Zharikova EI, Varaksin AA. Persistent and reparative neurogenesis in the juvenile masu salmon Oncorhynchus masou telencephalon after mechanical injury. Russ J Dev Biol 2017. [DOI: 10.1134/s106236041705006x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
|
22
|
Machado VM, Lourenço AS, Florindo C, Fernandes R, Carvalho CM, Araújo IM. Calpastatin Overexpression Preserves Cognitive Function Following Seizures, While Maintaining Post-Injury Neurogenesis. Front Mol Neurosci 2017; 10:60. [PMID: 28386216 PMCID: PMC5362605 DOI: 10.3389/fnmol.2017.00060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/22/2017] [Indexed: 01/27/2023] Open
Abstract
In the adult mammalian brain, new neurons continue to be produced throughout life in two main regions in the brain, the subgranular zone (SGZ) in the hippocampus and the subventricular zone in the walls of the lateral ventricles. Neural stem cells (NSCs) proliferate in these niches, and migrate as neuroblasts, to further differentiate in locations where new neurons are needed, either in normal or pathological conditions. However, the endogenous attempt of brain repair is not very efficient. Calpains are proteases known to be involved in neuronal damage and in cell proliferation, migration and differentiation of several cell types, though their effects on neurogenesis are not well known. Previous work by our group has shown that the absence of calpastatin (CAST), the endogenous inhibitor of calpains, impairs early stages of neurogenesis. Since the hippocampus is highly associated with learning and memory, we aimed to evaluate whether calpain inhibition would help improve cognitive recovery after lesion and efficiency of post-injury neurogenesis in this region. For that purpose, we used the kainic acid (KA) model of seizure-induced hippocampal lesion and mice overexpressing CAST. Selected cognitive tests were performed on the 3rd and 8th week after KA-induced lesion, and cell proliferation, migration and differentiation in the dentate gyrus (DG) of the hippocampus of adult mice were analyzed using specific markers. Cognitive recovery was evaluated by testing the animals for recognition, spatial and associative learning and memory. Cognitive function was preserved by CAST overexpression following seizures, while modulation of post-injury neurogenesis was similar to wild type (WT) mice. Calpain inhibition could still be potentially able to prevent the impairment in the formation of new neurons, given that the levels of calpain activity could be reduced under a certain threshold and other harmful effects from the pathological environment could also be controlled.
Collapse
Affiliation(s)
- Vanessa M Machado
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Center for Biomedical Research, CBMR, University of AlgarveFaro, Portugal; Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of AlgarveFaro, Portugal
| | - Ana Sofia Lourenço
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal; Center for Biomedical Research, CBMR, University of AlgarveFaro, Portugal; Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of AlgarveFaro, Portugal
| | - Cláudia Florindo
- Center for Biomedical Research, CBMR, University of AlgarveFaro, Portugal; Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of AlgarveFaro, Portugal
| | - Raquel Fernandes
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve Faro, Portugal
| | - Caetana M Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Inês M Araújo
- Center for Biomedical Research, CBMR, University of AlgarveFaro, Portugal; Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of AlgarveFaro, Portugal; Algarve Biomedical Center, University of AlgarveFaro, Portugal
| |
Collapse
|
23
|
Tian J, Luo Y, Chen W, Yang S, Wang H, Cui J, Lu Z, Lin Y, Bi Y. MeHg Suppressed Neuronal Potency of Hippocampal NSCs Contributing to the Puberal Spatial Memory Deficits. Biol Trace Elem Res 2016; 172:424-436. [PMID: 26743863 DOI: 10.1007/s12011-015-0609-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/23/2015] [Indexed: 12/21/2022]
Abstract
Hippocampal neurogenesis-related structural damage, particularly that leading to defective adult cognitive function, is considered an important risk factor for neurodegenerative and psychiatric diseases. Normal differentiation of neurons and glial cells during development is crucial in neurogenesis, which is particularly sensitive to the environmental toxicant methylmercury (MeHg). However, the exact effects of MeHg on hippocampal neural stem cell (hNSC) differentiation during puberty remain unknown. This study investigates whether MeHg exposure induces changes in hippocampal neurogenesis and whether these changes underlie cognitive defects in puberty. A rat model of methylmercury chloride (MeHgCl) exposure (0.4 mg/kg/day, PND 5-PND 33, 28 days) was established, and the Morris water maze was used to assess cognitive function. Primary hNSCs from hippocampal tissues of E16-day Sprague-Dawley rats were purified, identified, and cloned. hNSC proliferation and differentiation and the growth and morphology of newly generated neurons were observed by MTT and immunofluorescence assays. MeHg exposure induced defects in spatial learning and memory accompanied by a decrease in number of doublecortin (DCX)-positive cells in the dentate gyrus (DG). DCX is a surrogate marker for newly generated neurons. Proliferation and differentiation of hNSCs significantly decreased in the MeHg-treated groups. MeHg attenuated microtubule-associated protein-2 (MAP-2) expression in neurons and enhanced the glial fibrillary acidic protein (GFAP)-positive cell differentiation of hNSCs, thereby inducing degenerative changes in a dose-dependent manner. Moreover, MeHg induced deficits in hippocampus-dependent spatial learning and memory during adolescence as a consequence of decreased generation of DG neurons. Our findings suggested that MeHg exposure could be a potential risk factor for psychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jianying Tian
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
- Basic Medical School, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750004, China.
| | - Yougen Luo
- The Research Center of Neurodegenerative Diseases and Aging, Medical College of Jinggangshan University, Ji'an, Jiangxi, 343000, China
| | - Weiwei Chen
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Shengsen Yang
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Hao Wang
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Jing Cui
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Zhiyan Lu
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Yuanye Lin
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Yongyi Bi
- School of Public Health, Wuhan University, 115 Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
| |
Collapse
|
24
|
De la Rosa-Prieto C, Saiz-Sanchez D, Ubeda-Banon I, Flores-Cuadrado A, Martinez-Marcos A. Neurogenesis, Neurodegeneration, Interneuron Vulnerability, and Amyloid-β in the Olfactory Bulb of APP/PS1 Mouse Model of Alzheimer's Disease. Front Neurosci 2016; 10:227. [PMID: 27303258 PMCID: PMC4885141 DOI: 10.3389/fnins.2016.00227] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 05/06/2016] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, mostly idiopathic and with palliative treatment. Neuropathologically, it is characterized by intracellular neurofibrillary tangles of tau protein and extracellular plaques of amyloid β peptides. The relationship between AD and neurogenesis is unknown, but two facts are particularly relevant. First, early aggregation sites of both proteinopathies include the hippocampal formation and the olfactory bulb (OB), which have been correlated to memory and olfactory deficits, respectively. These areas are well-recognized integration zones of newly-born neurons in the adult brain. Second, molecules, such as amyloid precursor protein (APP) and presenilin-1 are common to both AD etiology and neurogenic development. Adult neurogenesis in AD models has been studied in the hippocampus, but only occasionally addressed in the OB and results are contradictory. To gain insight on the relationship between adult neurogenesis and AD, this work analyzes neurogenesis, neurodegeneration, interneuron vulnerability, and amyloid-β involvement in the OB of an AD model. Control and double-transgenic mice carrying the APP and the presenilin-1 genes, which give rise amyloid β plaques have been used. BrdU-treated animals have been studied at 16, 30, 43, and 56 weeks of age. New-born cell survival (BrdU), neuronal loss (using neuronal markers NeuN and PGP9.5), differential interneuron (calbindin-, parvalbumin-, calretinin- and somatostatin-expressing populations) vulnerability, and involvement by amyloid β have been analyzed. Neurogenesis increases with aging in the granule cell layer of control animals from 16 to 43 weeks. No neuronal loss has been observed after quantifying NeuN or PGP9.5. Regarding interneuron population vulnerability: calbindin-expressing neurons remains unchanged; parvalbumin-expressing neurons trend to increase with aging in transgenic animals; calretinin-expressing neurons increase with aging in transgenic mice and decrease in control animals and neurogenesis is higher in control as compared to transgenic animals at given ages, finally; somatostatin-expressing neurons of transgenic mice decrease with aging and as compared to controls. Amyloid β aggregates with aging in the granule cell layer, which may be related to the particular involvement of somatostatin-expressing cells.
Collapse
Affiliation(s)
- Carlos De la Rosa-Prieto
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Daniel Saiz-Sanchez
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Isabel Ubeda-Banon
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Alicia Flores-Cuadrado
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| | - Alino Martinez-Marcos
- Neuroplasticity and Neurodegeneration Laboratory, CRIB, Ciudad Real Medical School, Universidad de Castilla-La Mancha Ciudad Real, Spain
| |
Collapse
|
25
|
Ogino T, Sawada M, Takase H, Nakai C, Herranz-Pérez V, Cebrián-Silla A, Kaneko N, García-Verdugo JM, Sawamoto K. Characterization of multiciliated ependymal cells that emerge in the neurogenic niche of the aged zebrafish brain. J Comp Neurol 2016; 524:2982-92. [DOI: 10.1002/cne.24001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 02/24/2016] [Accepted: 03/15/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Takashi Ogino
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya 467-8601 Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya 467-8601 Japan
| | - Hiroshi Takase
- Core Laboratory; Nagoya City University Graduate School of Medical Sciences; Nagoya 467-8601 Japan
| | - Chiemi Nakai
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya 467-8601 Japan
| | - Vicente Herranz-Pérez
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles; Universidad de Valencia; CIBERNED 46980 Valencia Spain
- Unidad Mixta de Esclerosis Múltiple y Neurorregeneración; IIS Hospital La Fe; 46026 Valencia Spain
| | - Arantxa Cebrián-Silla
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles; Universidad de Valencia; CIBERNED 46980 Valencia Spain
| | - Naoko Kaneko
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya 467-8601 Japan
| | - José Manuel García-Verdugo
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles; Universidad de Valencia; CIBERNED 46980 Valencia Spain
- Unidad Mixta de Esclerosis Múltiple y Neurorregeneración; IIS Hospital La Fe; 46026 Valencia Spain
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya 467-8601 Japan
| |
Collapse
|
26
|
Capilla-Gonzalez V, Bonsu JM, Redmond KJ, Garcia-Verdugo JM, Quiñones-Hinojosa A. Implications of irradiating the subventricular zone stem cell niche. Stem Cell Res 2016; 16:387-96. [PMID: 26921873 PMCID: PMC8442998 DOI: 10.1016/j.scr.2016.02.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/10/2016] [Accepted: 02/14/2016] [Indexed: 01/19/2023] Open
Abstract
Radiation therapy is a standard treatment for brain tumor patients. However, it comes with side effects, such as neurological deficits. While likely multi-factorial, the effect may in part be associated with the impact of radiation on the neurogenic niches. In the adult mammalian brain, the neurogenic niches are localized in the subventricular zone (SVZ) of the lateral ventricles and the dentate gyrus of the hippocampus, where the neural stem cells (NSCs) reside. Several reports showed that radiation produces a drastic decrease in the proliferative capacity of these regions, which is related to functional decline. In particular, radiation to the SVZ led to a reduced long-term olfactory memory and a reduced capacity to respond to brain damage in animal models, as well as compromised tumor outcomes in patients. By contrast, other studies in humans suggested that increased radiation dose to the SVZ may be associated with longer progression-free survival in patients with high-grade glioma. In this review, we summarize the cellular and functional effects of irradiating the SVZ niche. In particular, we review the pros and cons of using radiation during brain tumor treatment, discussing the complex relationship between radiation dose to the SVZ and both tumor control and toxicity.
Collapse
Affiliation(s)
- Vivian Capilla-Gonzalez
- Department of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville 41092, Spain
| | - Janice M Bonsu
- Department of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Kristin J Redmond
- Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, University of Valencia, CIBERNED, Paterna 46980, Valencia, Spain
| | | |
Collapse
|
27
|
Hirota Y, Sawada M, Huang SH, Ogino T, Ohata S, Kubo A, Sawamoto K. Roles of Wnt Signaling in the Neurogenic Niche of the Adult Mouse Ventricular-Subventricular Zone. Neurochem Res 2015; 41:222-30. [PMID: 26572545 DOI: 10.1007/s11064-015-1766-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 01/08/2023]
Abstract
In many animal species, the production of new neurons (neurogenesis) occurs throughout life, in a specialized germinal region called the ventricular-subventricular zone (V-SVZ). In this region, neural stem cells undergo self-renewal and generate neural progenitor cells and new neurons. In the olfactory system, the new neurons migrate rostrally toward the olfactory bulb, where they differentiate into mature interneurons. V-SVZ-derived new neurons can also migrate toward sites of brain injury, where they contribute to neural regeneration. Recent studies indicate that two major branches of the Wnt signaling pathway, the Wnt/β-catenin and Wnt/planar cell polarity pathways, play essential roles in various facets of adult neurogenesis. Here, we review the Wnt signaling-mediated regulation of adult neurogenesis in the V-SVZ under physiological and pathological conditions.
Collapse
Affiliation(s)
- Yuki Hirota
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Shih-Hui Huang
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Takashi Ogino
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Shinya Ohata
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | - Akiharu Kubo
- Department of Dermatology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan.
| |
Collapse
|
28
|
Paredes MF, Sorrells SF, Garcia-Verdugo JM, Alvarez-Buylla A. Brain size and limits to adult neurogenesis. J Comp Neurol 2015; 524:646-64. [PMID: 26417888 DOI: 10.1002/cne.23896] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 08/28/2015] [Accepted: 09/08/2015] [Indexed: 12/31/2022]
Abstract
The walls of the cerebral ventricles in the developing embryo harbor the primary neural stem cells from which most neurons and glia derive. In many vertebrates, neurogenesis continues postnatally and into adulthood in this region. Adult neurogenesis at the ventricle has been most extensively studied in organisms with small brains, such as reptiles, birds, and rodents. In reptiles and birds, these progenitor cells give rise to young neurons that migrate into many regions of the forebrain. Neurogenesis in adult rodents is also relatively widespread along the lateral ventricles, but migration is largely restricted to the rostral migratory stream into the olfactory bulb. Recent work indicates that the wall of the lateral ventricle is highly regionalized, with progenitor cells giving rise to different types of neurons depending on their location. In species with larger brains, young neurons born in these spatially specified domains become dramatically separated from potential final destinations. Here we hypothesize that the increase in size and topographical complexity (e.g., intervening white matter tracts) in larger brains may severely limit the long-term contribution of new neurons born close to, or in, the ventricular wall. We compare the process of adult neuronal birth, migration, and integration across species with different brain sizes, and discuss how early regional specification of progenitor cells may interact with brain size and affect where and when new neurons are added.
Collapse
Affiliation(s)
- Mercedes F Paredes
- Department of Neurological Surgery, University of California, San Francisco, CA, 94143, USA
| | - Shawn F Sorrells
- Department of Neurological Surgery, University of California, San Francisco, CA, 94143, USA.,University of California, San Francisco, CA, 94143, USA
| | - Jose M Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universidad de Valencia, CIBERNED, 46980 Valencia, Spain
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
29
|
Machado VM, Morte MI, Carreira BP, Azevedo MM, Takano J, Iwata N, Saido TC, Asmussen H, Horwitz AR, Carvalho CM, Araújo IM. Involvement of calpains in adult neurogenesis: implications for stroke. Front Cell Neurosci 2015; 9:22. [PMID: 25698931 PMCID: PMC4316774 DOI: 10.3389/fncel.2015.00022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/13/2015] [Indexed: 11/13/2022] Open
Abstract
Calpains are ubiquitous proteases involved in cell proliferation, adhesion and motility. In the brain, calpains have been associated with neuronal damage in both acute and neurodegenerative disorders, but their physiological function in the nervous system remains elusive. During brain ischemia, there is a large increase in the levels of intracellular calcium, leading to the activation of calpains. Inhibition of these proteases has been shown to reduce neuronal death in a variety of stroke models. On the other hand, after stroke, neural stem cells (NSC) increase their proliferation and newly formed neuroblasts migrate towards the site of injury. However, the process of forming new neurons after injury is not efficient and finding ways to improve it may help with recovery after lesion. Understanding the role of calpains in the process of neurogenesis may therefore open a new window for the treatment of stroke. We investigated the involvement of calpains in NSC proliferation and neuroblast migration in two highly neurogenic regions in the mouse brain, the dentate gyrus (DG) and the subventricular zone (SVZ). We used mice that lack calpastatin, the endogenous calpain inhibitor, and calpains were also modulated directly, using calpeptin, a pharmacological calpain inhibitor. Calpastatin deletion impaired both NSC proliferation and neuroblast migration. Calpain inhibition increased NSC proliferation, migration speed and migration distance in cells from the SVZ. Overall, our work suggests that calpains are important for neurogenesis and encourages further research on their neurogenic role. Prospective therapies targeting calpain activity may improve the formation of new neurons following stroke, in addition to affording neuroprotection.
Collapse
Affiliation(s)
- Vanessa M Machado
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve Faro, Portugal ; IBB-Institute for Biotechnology and Bioengineering, Center for Molecular and Structural Biomedicine, University of Algarve Faro, Portugal ; Center for Biomedical Research, CBMR, University of Algarve Faro, Portugal ; Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Maria I Morte
- Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Bruno P Carreira
- Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Maria M Azevedo
- Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Jiro Takano
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute Wako-shi, Saitama, Japan
| | - Nobuhisa Iwata
- Graduate School of Biomedical Sciences, Nagasaki University Nagasaki, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute Wako-shi, Saitama, Japan
| | - Hannelore Asmussen
- Department of Cell Biology, University of Virginia School of Medicine Charlottesville, VA, USA
| | - Alan R Horwitz
- Department of Cell Biology, University of Virginia School of Medicine Charlottesville, VA, USA
| | - Caetana M Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| | - Inês M Araújo
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve Faro, Portugal ; IBB-Institute for Biotechnology and Bioengineering, Center for Molecular and Structural Biomedicine, University of Algarve Faro, Portugal ; Center for Biomedical Research, CBMR, University of Algarve Faro, Portugal ; Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal
| |
Collapse
|
30
|
McAllister TW. Genetic factors in traumatic brain injury. HANDBOOK OF CLINICAL NEUROLOGY 2015; 128:723-39. [DOI: 10.1016/b978-0-444-63521-1.00045-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
31
|
Fan C, Zhang M, Shang L, Cynthia NA, Li Z, Yang Z, Chen D, Huang J, Xiong K. Short-term environmental enrichment exposure induces proliferation and maturation of doublecortin-positive cells in the prefrontal cortex. Neural Regen Res 2014; 9:318-28. [PMID: 25206818 PMCID: PMC4146142 DOI: 10.4103/1673-5374.128231] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2013] [Indexed: 11/04/2022] Open
Abstract
Previous studies have demonstrated that doublecortin-positive immature neurons exist predominantly in the superficial layer of the cerebral cortex of adult mammals such as guinea pigs, and these neurons exhibit very weak properties of self-proliferation during adulthood under physiological conditions. To verify whether environmental enrichment has an impact on the proliferation and maturation of these immature neurons in the prefrontal cortex of adult guinea pigs, healthy adult guinea pigs were subjected to short-term environmental enrichment. Animals were allowed to play with various cognitive and physical stimulating objects over a period of 2 weeks, twice per day, for 60 minutes each. Immunofluorescence staining results indicated that the number of doublecortin-positive cells in layer II of the prefrontal cortex was significantly increased after short-term environmental enrichment exposure. In addition, these doublecortin-positive cells co-expressed 5-bromo-2-deoxyuridine (a marker of cell proliferation), c-Fos (a marker of cell viability) and NeuN (a marker of mature neurons). Experimental findings showed that short-term environmental enrichment can induce proliferation, activation and maturation of doublecortin-positive cells in layer II of the prefrontal cortex of adult guinea pigs.
Collapse
Affiliation(s)
- Chunling Fan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan Province, China
| | - Mengqi Zhang
- Grade 2006, Eight-year Medicine Doctor Program, Central South University Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Lei Shang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan Province, China
| | - Ngobe Akume Cynthia
- Grade 2011, Six-year Medicine Program of International Student, Central South University Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Zhi Li
- Grade 2008, Eight-year Medicine Doctor Program, Central South University Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Zhenyu Yang
- Grade 2008, Eight-year Medicine Doctor Program, Central South University Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan Province, China
| | - Jufang Huang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan Province, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan Province, China
| |
Collapse
|
32
|
Yan Y, Ma T, Gong K, Ao Q, Zhang X, Gong Y. Adipose-derived mesenchymal stem cell transplantation promotes adult neurogenesis in the brains of Alzheimer's disease mice. Neural Regen Res 2014; 9:798-805. [PMID: 25206892 PMCID: PMC4146257 DOI: 10.4103/1673-5374.131596] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2014] [Indexed: 12/11/2022] Open
Abstract
In the present study, we transplanted adipose-derived mesenchymal stem cells into the hippocampi of APP/PS1 transgenic Alzheimer's disease model mice. Immunofluorescence staining revealed that the number of newly generated (BrdU(+)) cells in the subgranular zone of the dentate gyrus in the hippocampus was significantly higher in Alzheimer's disease mice after adipose-derived mesenchymal stem cell transplantation, and there was also a significant increase in the number of BrdU(+)/DCX(+) neuroblasts in these animals. Adipose-derived mesenchymal stem cell transplantation enhanced neurogenic activity in the subventricular zone as well. Furthermore, adipose-derived mesenchymal stem cell transplantation reduced oxidative stress and alleviated cognitive impairment in the mice. Based on these findings, we propose that adipose-derived mesenchymal stem cell transplantation enhances endogenous neurogenesis in both the subgranular and subventricular zones in APP/PS1 transgenic Alzheimer's disease mice, thereby facilitating functional recovery.
Collapse
Affiliation(s)
- Yufang Yan
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tuo Ma
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Kai Gong
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiang Ao
- Institute of Neurological Disorders, Yuquan Hospital, Tsinghua University, Beijing, China
| | - Xiufang Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yandao Gong
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
33
|
Activated CD8+ T lymphocytes inhibit neural stem/progenitor cell proliferation: role of interferon-gamma. PLoS One 2014; 9:e105219. [PMID: 25133679 PMCID: PMC4136865 DOI: 10.1371/journal.pone.0105219] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 07/21/2014] [Indexed: 12/03/2022] Open
Abstract
The ability of neural stem/progenitor cells (NSCs) to self-renew, migrate to damaged sites, and differentiate into neurons has renewed interest in using them in therapies for neurodegenerative disorders. Neurological diseases, including viral infections of the brain, are often accompanied by chronic inflammation, whose impact on NSC function remains unexplored. We have previously shown that chronic neuroinflammation, a hallmark of experimental herpes simplex encephalitis (HSE) in mice, is dominated by brain-infiltrating activated CD8 T-cells. In the present study, activated CD8 lymphocytes were found to suppress NSC proliferation profoundly. Luciferase positive (luc+) NSCs co-cultured with activated, MHC-matched, CD8+ lymphocytes (luc−) showed two- to five-fold lower luminescence than co-cultures with un-stimulated lymphocytes. On the other hand, similarly activated CD4+ lymphocytes did not suppress NSC growth. This differential lymphocyte effect on proliferation was confirmed by decreased BrdU uptake by NSC cultured with activated CD8 T-cells. Interestingly, neutralizing antibodies to interferon-gamma (IFN-γ) reversed the impact of CD8 lymphocytes on NSCs. Antibodies specific to the IFN-γ receptor-1 subunit complex abrogated the inhibitory effects of both CD8 lymphocytes and IFN-γ, indicating that the inhibitory effect of these cells was mediated by IFN-γ in a receptor-specific manner. In addition, activated CD8 lymphocytes decreased levels of nestin and Sox2 expression in NSCs while increasing GFAP expression, suggesting possible induction of an altered differentiation state. Furthermore, NSCs obtained from IFN-γ receptor-1 knock-out embryos were refractory to the inhibitory effects of activated CD8+ T lymphocytes on cell proliferation and Sox2 expression. Taken together, the studies presented here demonstrate a role for activated CD8 T-cells in regulating NSC function mediated through the production of IFN-γ. This cytokine may influence neuro-restorative processes and ultimately contribute to the long-term sequelae commonly seen following herpes encephalitis.
Collapse
|
34
|
Fitzsimons CP, van Bodegraven E, Schouten M, Lardenoije R, Kompotis K, Kenis G, van den Hurk M, Boks MP, Biojone C, Joca S, Steinbusch HWM, Lunnon K, Mastroeni DF, Mill J, Lucassen PJ, Coleman PD, van den Hove DLA, Rutten BPF. Epigenetic regulation of adult neural stem cells: implications for Alzheimer's disease. Mol Neurodegener 2014; 9:25. [PMID: 24964731 PMCID: PMC4080757 DOI: 10.1186/1750-1326-9-25] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/06/2014] [Indexed: 01/27/2023] Open
Abstract
Experimental evidence has demonstrated that several aspects of adult neural stem cells (NSCs), including their quiescence, proliferation, fate specification and differentiation, are regulated by epigenetic mechanisms. These control the expression of specific sets of genes, often including those encoding for small non-coding RNAs, indicating a complex interplay between various epigenetic factors and cellular functions.Previous studies had indicated that in addition to the neuropathology in Alzheimer's disease (AD), plasticity-related changes are observed in brain areas with ongoing neurogenesis, like the hippocampus and subventricular zone. Given the role of stem cells e.g. in hippocampal functions like cognition, and given their potential for brain repair, we here review the epigenetic mechanisms relevant for NSCs and AD etiology. Understanding the molecular mechanisms involved in the epigenetic regulation of adult NSCs will advance our knowledge on the role of adult neurogenesis in degeneration and possibly regeneration in the AD brain.
Collapse
Affiliation(s)
- Carlos P Fitzsimons
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Emma van Bodegraven
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Marijn Schouten
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Roy Lardenoije
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Konstantinos Kompotis
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Gunter Kenis
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Mark van den Hurk
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Marco P Boks
- Department Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Biojone
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Samia Joca
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Harry WM Steinbusch
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Katie Lunnon
- University of Exeter Medical School, RILD Level 4, Barrack Road, University of Exeter, Devon, UK
| | - Diego F Mastroeni
- University of Exeter Medical School, RILD Level 4, Barrack Road, University of Exeter, Devon, UK
| | - Jonathan Mill
- University of Exeter Medical School, RILD Level 4, Barrack Road, University of Exeter, Devon, UK
| | - Paul J Lucassen
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Paul D Coleman
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Daniel LA van den Hove
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Bart PF Rutten
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
35
|
Eiriz MF, Valero J, Malva JO, Bernardino L. New insights into the role of histamine in subventricular zone-olfactory bulb neurogenesis. Front Neurosci 2014; 8:142. [PMID: 24982610 PMCID: PMC4058902 DOI: 10.3389/fnins.2014.00142] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/21/2014] [Indexed: 11/21/2022] Open
Abstract
The subventricular zone (SVZ) contains neural stem cells (NSCs) that generate new neurons throughout life. Many brain diseases stimulate NSCs proliferation, neuronal differentiation and homing of these newborns cells into damaged regions. However, complete cell replacement has never been fully achieved. Hence, the identification of proneurogenic factors crucial for stem cell-based therapies will have an impact in brain repair. Histamine, a neurotransmitter and immune mediator, has been recently described to modulate proliferation and commitment of NSCs. Histamine levels are increased in the brain parenchyma and at the cerebrospinal fluid (CSF) upon inflammation and brain injury, thus being able to modulate neurogenesis. Herein, we add new data showing that in vivo administration of histamine in the lateral ventricles has a potent proneurogenic effect, increasing the production of new neuroblasts in the SVZ that ultimately reach the olfactory bulb (OB). This report emphasizes the multidimensional effects of histamine in the modulation of NSCs dynamics and sheds light into the promising therapeutic role of histamine for brain regenerative medicine.
Collapse
Affiliation(s)
- Maria F Eiriz
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra (CNC-UC) Coimbra, Portugal
| | - Jorge Valero
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra (CNC-UC) Coimbra, Portugal
| | - João O Malva
- Faculty of Medicine, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra Coimbra, Portugal
| | - Liliana Bernardino
- Faculty of Health Sciences, Health Sciences Research Center, University of Beira Interior Covilhã, Portugal
| |
Collapse
|
36
|
Enomoto A, Asai N, Takahashi M. [Mechanisms for the differentiation of postnatal and adult neural stem cells: new insights and pathological issues based on the analysis of Girdin]. Nihon Yakurigaku Zasshi 2014; 143:289-294. [PMID: 24919555 DOI: 10.1254/fpj.143.289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
|
37
|
Sawada M, Matsumoto M, Sawamoto K. Vascular regulation of adult neurogenesis under physiological and pathological conditions. Front Neurosci 2014; 8:53. [PMID: 24672424 PMCID: PMC3955849 DOI: 10.3389/fnins.2014.00053] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 02/26/2014] [Indexed: 01/16/2023] Open
Abstract
Neural stem cells in the mammalian adult brain continuously produce new neurons throughout life. Accumulating evidence in rodents suggests that various aspects of adult neurogenesis, including the genesis, migration, and maturation of new neurons, are regulated by factors derived from blood vessels and their microenvironment. Brain injury enhances both neurogenesis and angiogenesis, thereby promoting the cooperative regeneration of neurons and blood vessels. In this paper, we briefly review the mechanisms for the vascular regulation of adult neurogenesis in the ventricular-subventricular zone under physiological and pathological conditions, and discuss their clinical potential for brain regeneration strategies.
Collapse
Affiliation(s)
- Masato Sawada
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| | - Mami Matsumoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| |
Collapse
|
38
|
Varela-Nallar L, Rojas-Abalos M, Abbott AC, Moya EA, Iturriaga R, Inestrosa NC. Chronic hypoxia induces the activation of the Wnt/β-catenin signaling pathway and stimulates hippocampal neurogenesis in wild-type and APPswe-PS1ΔE9 transgenic mice in vivo. Front Cell Neurosci 2014; 8:17. [PMID: 24574965 PMCID: PMC3918655 DOI: 10.3389/fncel.2014.00017] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/10/2014] [Indexed: 01/01/2023] Open
Abstract
Hypoxia modulates proliferation and differentiation of cultured embryonic and adult stem cells, an effect that includes β-catenin, a key component of the canonical Wnt signaling pathway. Here we studied the effect of mild hypoxia on the activity of the Wnt/β-catenin signaling pathway in the hippocampus of adult mice in vivo. The hypoxia-inducible transcription factor-1α (HIF-1α) was analyzed as a molecular control of the physiological hypoxic response. Exposure to chronic hypoxia (10% oxygen for 6–72 h) stimulated the activation of the Wnt/β-catenin signaling pathway. Because the Wnt/β-catenin pathway is a positive modulator of adult neurogenesis, we evaluated whether chronic hypoxia was able to stimulate neurogenesis in the subgranular zone (SGZ) of the hippocampal dentate gyrus. Results indicate that hypoxia increased cell proliferation and neurogenesis in adult wild-type mice as determined by Ki67 staining, Bromodeoxyuridine (BrdU) incorporation and double labeling with doublecortin (DCX). Chronic hypoxia also induced neurogenesis in a double transgenic APPswe-PS1ΔE9 mouse model of Alzheimer’s disease (AD), which shows decreased levels of neurogenesis in the SGZ. Our results show for the first time that exposure to hypoxia in vivo can induce the activation of the Wnt/β-catenin signaling cascade in the hippocampus, suggesting that mild hypoxia may have a therapeutic value in neurodegenerative disorders associated with altered Wnt signaling in the brain and also in pathological conditions in which hippocampal neurogenesis is impaired.
Collapse
Affiliation(s)
- Lorena Varela-Nallar
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile ; Centro de Investigaciones Biomédicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello Santiago, Chile
| | - Macarena Rojas-Abalos
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Ana C Abbott
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Esteban A Moya
- Laboratorio de Neurobiología, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
39
|
White matter and SVZ serve as endogenous sources of glial progenitor cells for self-repair in neonatal rats with ischemic PVL. Brain Res 2013; 1535:38-51. [DOI: 10.1016/j.brainres.2013.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 07/31/2013] [Accepted: 08/04/2013] [Indexed: 01/18/2023]
|
40
|
Morte MI, Carreira BP, Machado V, Carmo A, Nunes-Correia I, Carvalho CM, Araújo IM. Evaluation of proliferation of neural stem cells in vitro and in vivo. ACTA ACUST UNITED AC 2013; Chapter 2:Unit 2D.14. [PMID: 23404673 DOI: 10.1002/9780470151808.sc02d14s24] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
This unit describes two basic protocols for the detection of the proliferation of neural stem cells (NSC). The first one addresses cell proliferation in cultures, starting with primary cell cultures isolated from the mouse subventricular zone (SVZ), in which SVZ-derived NSC are kept in culture as neurospheres. By using this culture system, we are able to study different stages of adult neurogenesis, such as proliferation, differentiation, migration, and survival. Thus, in the first basic protocol, we describe two different techniques to evaluate cell proliferation based on EdU incorporation: (a) immunocytochemistry and (b) flow cytometry. EdU, a new thymidine analog, which is detected by a reproducible and sensitive method based on click chemistry, does not require DNA denaturation, as is the case with BrdU. Thus, co-labeling of EdU with other specific antibodies of extracellular or intracellular targets, as well as other DNA dyes, is possible. In the second basic protocol, we describe an in vivo assay to evaluate proliferation of NSC in the dentate gyrus of hippocampus of adult mice, by both BrdU and EdU detection. With this approach, it is also possible to study different stages of adult neurogenesis, by co-labeling thymidine analogs with other specific markers, such as doublecortin (DCX) or neuronal nuclei protein (NeuN).
Collapse
Affiliation(s)
- Maria Inês Morte
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | |
Collapse
|
41
|
Shu HF, Kuang YQ, Liu SY, Yu SX, Zhang CQ, Zheng DH, Gu JW, Yang H. Endogenous subventricular zone neural progenitors contribute to the formation and hyperexcitability of experimental model of focal microgyria. J Mol Neurosci 2013; 52:586-97. [PMID: 24057922 DOI: 10.1007/s12031-013-0114-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 09/02/2013] [Indexed: 12/30/2022]
Abstract
Microgyria is associated with epilepsy and due to developmental disruption of neuronal migration. However, the role of endogenous subventricular zone-derived neural progenitors (SDNPs) in formation and hyperexcitability has not been fully elucidated. Here, we establish a neonatal cortex freeze-lesion (FL) model, which was considered as a model for focal microgyria, and simultaneously label SDNPs by CM-DiI. Morphological investigation showed that SDNPs migrated into FL and differentiated into neuronal and glia cell types, suggesting the involvement of endogenous SDNPs in the formation of FL-induced microgyria. Patch-clamp recordings in CM-DiI positive (CM-DiI(+)) pyramidal neurons within FL indicated an increase in frequency of spontaneous action potentials, while the resting membrane potential did not differ from the controls. We also found that spontaneous excitatory postsynaptic currents (EPSCs) increased in frequency but not in amplitude compared with controls. The evoked EPSCs showed a significant increase of 10-90% in rise time and decay time in the CM-DiI(+) neurons. Moreover, paired-pulse facilitation was dramatically larger in CM-DiI(+) pyramidal neurons. Western blotting data showed that AMPA and NMDA receptors were increased to some extent in the FL cortex compared with controls, and the NMDA/AMPA ratio of eEPSCs at CM-DiI(+) pyramidal neurons was significantly increased. Taken together, our findings provide novel evidence for the contribution of endogenous SDNPs in the formation and epileptogenicity of FL-induced focal microgyria.
Collapse
Affiliation(s)
- Hai-Feng Shu
- Department of Neurosurgery, General Hospital of the People's Liberation Army Chengdu Military Region, Sichuan, 610000, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wu MD, Montgomery SL, Rivera-Escalera F, Olschowka JA, O’Banion MK. Sustained IL-1β expression impairs adult hippocampal neurogenesis independent of IL-1 signaling in nestin+ neural precursor cells. Brain Behav Immun 2013; 32:9-18. [PMID: 23510988 PMCID: PMC3686979 DOI: 10.1016/j.bbi.2013.03.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 02/18/2013] [Accepted: 03/07/2013] [Indexed: 01/17/2023] Open
Abstract
Alterations in adult hippocampal neurogenesis have been observed in numerous neurological diseases that contain a neuroinflammatory component. Interleukin-1β (IL-1β) is a pro-inflammatory cytokine that contributes to neuroinflammation in many CNS disorders. Our previous results reveal a severe reduction in adult hippocampal neurogenesis due to focal and chronic expression of IL-1β in a transgenic mouse model, IL-1β(XAT), that evokes a complex neuroinflammatory response. Other investigators have shown that IL-1β can bind directly to neural precursors to cause cell cycle arrest in vitro. In order to observe if IL-1 signaling is necessary in vivo, we conditionally knocked out MyD88, an adapter protein essential for IL-1 signaling, in nestin(+) neural precursor cells (NPCs) in the presence of IL-1β-dependent inflammation. Our results show that conditional knockout of MyD88 does not prevent IL-1β-induced reduction in neuroblasts using a genetic fate mapping model. Interestingly, MyD88 deficiency in nestin(+) NPCs causes an increase in the number of astrocytes in the presence of IL-1β, suggesting that MyD88-dependent signaling is important in limiting astroglial differentiation due to inflammation. MyD88 deficiency does not alter the fate of NPCs in the absence of inflammation. Furthermore, the inflammatory milieu due to IL-1β is not affected by the absence of MyD88 in nestin(+) NPCs. These results show that sustained IL-1β causes a reduction in adult hippocampal neurogenesis that is independent of MyD88-dependent signaling in nestin(+) NPCs, suggesting an indirect negative effect of IL-1β on neurogenesis.
Collapse
Affiliation(s)
| | | | | | | | - M. Kerry O’Banion
- Corresponding Author: M. Kerry O’Banion, MD, PhD, 601 Elmwood Avenue, Box 603, University of Rochester Medical Center, Rochester, NY 14642, Phone: 585 275-5185, FAX: 585 756-5334,
| |
Collapse
|
43
|
Kako E, Kaneko N, Aoyama M, Hida H, Takebayashi H, Ikenaka K, Asai K, Togari H, Sobue K, Sawamoto K. Subventricular zone-derived oligodendrogenesis in injured neonatal white matter in mice enhanced by a nonerythropoietic erythropoietin derivative. Stem Cells 2013; 30:2234-47. [PMID: 22890889 DOI: 10.1002/stem.1202] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Perinatal hypoxia-ischemia (HI) frequently causes white-matter injury, leading to severe neurological deficits and mortality, and only limited therapeutic options exist. The white matter of animal models and human patients with HI-induced brain injury contains increased numbers of oligodendrocyte progenitor cells (OPCs). However, the origin and fates of these OPCs and their potential to repair injured white matter remain unclear. Here, using cell-type- and region-specific genetic labeling methods in a mouse HI model, we characterized the Olig2-expressing OPCs. We found that after HI, Olig2+ cells increased in the posterior part of the subventricular zone (pSVZ) and migrated into the injured white matter. However, their oligodendrocytic differentiation efficiency was severely compromised compared with the OPCs in normal tissue, indicating the need for an intervention to promote their differentiation. Erythropoietin (EPO) treatment is a promising candidate, but it has detrimental effects that preclude its clinical use for brain injury. We found that long-term postinjury treatment with a nonerythropoietic derivative of EPO, asialo-erythropoietin, promoted the maturation of pSVZ-derived OPCs and the recovery of neurological function, without affecting hematopoiesis. These results demonstrate the limitation and potential of endogenous OPCs in the pSVZ as a therapeutic target for treating neonatal white-matter injury.
Collapse
Affiliation(s)
- Eisuke Kako
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Carnicero E, Alonso M, Carretero R, Lamus F, Moro J, de la Mano A, Fernández J, Gato A. Embryonic Cerebrospinal Fluid Activates Neurogenesis of Neural Precursors within the Subventricular Zone of the Adult Mouse Brain. Cells Tissues Organs 2013; 198:398-404. [DOI: 10.1159/000356983] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2013] [Indexed: 11/19/2022] Open
|
45
|
Yang P, Zhang J, Zhao L, Jiao Q, Jin H, Xiao X, Zhang H, Hu M, Lu H, Liu Y. Developmental distribution pattern of metabotropic glutamate receptor 5 in prenatal human hippocampus. Neurosci Bull 2012; 28:704-14. [PMID: 23225313 DOI: 10.1007/s12264-012-1286-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 05/17/2012] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Metabotropic glutamate receptor 5 (mGluR5) is concentrated in zones of active neurogenesis in the prenatal and postnatal rodent brain and plays an important role in the regulation of neurogenesis. However, little is known about mGluR5 in the prenatal human brain. Here, we aimed to explore the expression pattern and cellular distribution of mGluR5 in human fetal hippocampus. METHODS Thirty-four human fetuses were divided into four groups according to gestational age: 9-11, 14-16, 22-24 and 32-36 weeks. The hippocampus was dissected out and prepared. The protein and mRNA expression of mGluR5 were evaluated by Western blot and immunohistochemistry or real-time PCR. The cellular distribution of mGluR5 was observed with double-labeling immunofluorescence. RESULTS Both mGluR5 mRNA and protein were detected in the prenatal human hippocampus by real-time PCR and immunoblotting, and the expression levels increased gradually over time. The immunohistochemistry results were consistent with immunoblotting and showed that mGluR5 immunoreactivity was mainly present in the inner marginal zone (IMZ), hippocampal plate (HP) and ventricular zone (VZ). The double-labeling immunofluorescence showed that mGluR5 was present in neural stem cells (nestin-positive), neuroblasts (DCX-positive) and mature neurons (NeuN-positive), but not in typical astrocytes (GFAP-positive). The cells co-expressing mGluR5 and nestin were mainly located in the IMZ, HP and subplate at 11 weeks, all layers at 16 weeks, and CA1 at 24 weeks. As development proceeded, the number of mGluR5/nestin double-positive cells decreased gradually so that there were only a handful of double-labeled cells at 32 weeks. However, mGluR5/DCX double-positive cells were only found in the HP, IZ and IMZ at 11 weeks. CONCLUSION The pattern of mGluR5 expression by neural stem/progenitor cells, neuroblasts and neurons provides important anatomical evidence for the role of mGluR5 in the regulation of human hippocampal development.
Collapse
Affiliation(s)
- Pengbo Yang
- Institute of Neurobiology, Key Laboratory for Environment and Genes Related to Diseases of the Ministry of Education, Xi'an Jiaotong University College of Medicine, Xi'an 710061, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Growth factors released from gelatin hydrogel microspheres increase new neurons in the adult mouse brain. Stem Cells Int 2012; 2012:915160. [PMID: 23093979 PMCID: PMC3474987 DOI: 10.1155/2012/915160] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 08/09/2012] [Indexed: 11/18/2022] Open
Abstract
Recent studies have shown that new neurons are continuously generated by endogenous neural stem cells in the subventricular zone (SVZ) of the adult mammalian brain. Some of these new neurons migrate to injured brain tissues and differentiate into mature neurons, suggesting that such new neurons may be able to replace neurons lost to degenerative disease or injury and improve or repair neurological deficits. Here, we tested whether delivering growth factors via gelatin hydrogel microspheres would support neurogenesis in the SVZ. Insulin-like growth factor-1 (IGF-1)-containing microspheres increased the number of new neurons in the SVZ. Hepatocyte growth factor (HGF)-containing microspheres increased the number of new neurons migrating from the SVZ towards the injured striatum in a stroke model in mouse. These results suggest that the strategy of using gelatin hydrogel microspheres to achieve the sustained release of growth factors holds promise for the clinical regeneration of damaged brain tissues from endogenous neural stem cells in the adult SVZ.
Collapse
|
47
|
Kishimoto N, Alfaro-Cervello C, Shimizu K, Asakawa K, Urasaki A, Nonaka S, Kawakami K, Garcia-Verdugo JM, Sawamoto K. Migration of neuronal precursors from the telencephalic ventricular zone into the olfactory bulb in adult zebrafish. J Comp Neurol 2012; 519:3549-65. [PMID: 21800305 DOI: 10.1002/cne.22722] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the brain of adult mammals, neuronal precursors are generated in the subventricular zone in the lateral wall of the lateral ventricles and migrate into the olfactory bulbs (OBs) through a well-studied route called the rostral migratory stream (RMS). Recent studies have revealed that a comparable neural stem cell niche is widely conserved at the ventricular wall of adult vertebrates. However, little is known about the migration route of neuronal precursors in nonmammalian adult brains. Here, we show that, in the adult zebrafish, a cluster of neuronal precursors generated in the telencephalic ventricular zone migrates into the OB via a route equivalent to the mammalian RMS. Unlike the mammalian RMS, these neuronal precursors are not surrounded by glial tubes, although radial glial cells with a single cilium lined the telencephalic ventricular wall, much as in embryonic and neonatal mammals. To observe the migrating neuronal precursors in living brain tissue, we established a brain hemisphere culture using a zebrafish line carrying a GFP transgene driven by the neurogenin1 (ngn1) promoter. In these fish, GFP was observed in the neuronal precursors migrating in the RMS, some of which were aligned with blood vessels. Numerous ngn1:gfp-positive cells were observed migrating tangentially in the RMS-like route medial to the OB. Taken together, our results suggest that the RMS in the adult zebrafish telencephalon is a functional migratory pathway. This is the first evidence for the tangential migration of neuronal precursors in a nonmammalian adult telencephalon.
Collapse
Affiliation(s)
- Norihito Kishimoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Neural stem cell specific fluorescent chemical probe binding to FABP7. Proc Natl Acad Sci U S A 2012; 109:10214-7. [PMID: 22689954 DOI: 10.1073/pnas.1200817109] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fluorescent small molecules have become indispensable tools for biomedical research along with the rapidly developing optical imaging technology. We report here a neural stem cell specific boron-dipyrromethane (BODIPY) derivative compound of designation red 3 (CDr3), developed through a high throughput/content screening of in-house generated diversity oriented fluorescence library in stem cells at different developmental stages. This novel compound specifically detects living neural stem cells of both human and mouse origin. Furthermore, we identified its binding target by proteomic analysis as fatty acid binding protein 7 (FABP7), also known as brain lipid binding protein) which is highly expressed in neural stem cells and localized in the cytoplasm. CDr3 will be a valuable chemical tool in the study and applications of neural stem cells.
Collapse
|
49
|
More SV, Koppula S, Kim BW, Choi DK. The role of bioactive compounds on the promotion of neurite outgrowth. Molecules 2012; 17:6728-53. [PMID: 22664464 PMCID: PMC6268652 DOI: 10.3390/molecules17066728] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/01/2012] [Accepted: 05/28/2012] [Indexed: 12/18/2022] Open
Abstract
Neurite loss is one of the cardinal features of neuronal injury. Apart from neuroprotection, reorganization of the lost neuronal network in the injured brain is necessary for the restoration of normal physiological functions. Neuritogenic activity of endogenous molecules in the brain such as nerve growth factor is well documented and supported by scientific studies which show innumerable compounds having neurite outgrowth activity from natural sources. Since the damaged brain lacks the reconstructive capacity, more efforts in research are focused on the identification of compounds that promote the reformation of neuronal networks. An abundancy of natural resources along with the corresponding activity profiles have shown promising results in the field of neuroscience. Recently, importance has also been placed on understanding neurite formation by natural products in relation to neuronal injury. Arrays of natural herbal products having plentiful active constituents have been found to enhance neurite outgrowth. They act synergistically with neurotrophic factors to promote neuritogenesis in the diseased brain. Therefore use of natural products for neuroregeneration provides new insights in drug development for treating neuronal injury. In this study, various compounds from natural sources with potential neurite outgrowth activity are reviewed in experimental models.
Collapse
Affiliation(s)
| | | | | | - Dong-Kug Choi
- Department of Biotechnology, Research Institute for Biomedical and Health Science, Konkuk University, Chungju 380-701, Korea
| |
Collapse
|
50
|
Putative roles for phospholipase Cη enzymes in neuronal Ca2+ signal modulation. Biochem Soc Trans 2012; 40:282-6. [PMID: 22260706 DOI: 10.1042/bst20110622] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The most recently identified PLC (phospholipase C) enzymes belong to the PLCη family. Their unique Ca2+-sensitivity and their specific appearance in neurons have attracted great attention since their discovery; however, their physiological role(s) in neurons are still yet to be established. PLCη enzymes are expressed in the neocortex, hippocampus and cerebellum. PLCη2 is also expressed at high levels in pituitary gland, pineal gland and in the retina. Driven by the specific localization of PLCη enzymes in different brain areas, in the present paper, we discuss the roles that they may play in neural processes, including differentiation, memory formation, circadian rhythm regulation, neurotransmitter/hormone release and the pathogenesis of neurodegenerative disorders associated with aberrant Ca2+ signalling, such as Alzheimer's disease.
Collapse
|