1
|
Nabeel Mustafa A, Salih Mahdi M, Ballal S, Chahar M, Verma R, Ali Al-Nuaimi AM, Kumar MR, Kadhim A Al-Hussein R, Adil M, Jasem Jawad M. Netrin-1: Key insights in neural development and disorders. Tissue Cell 2025; 93:102678. [PMID: 39719818 DOI: 10.1016/j.tice.2024.102678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/26/2024]
Abstract
Netrin-1, an essential extracellular protein, has gained significant attention due to its pivotal role in guiding axon and cell migration during embryonic development. The fundamental significance of netrin-1 in developmental biology is reflected in its high conservation across different species as a part of the netrin family. The bifunctional nature of netrin-1 demonstrates its functional versatility, as it can function as either a repellent or an attractant according to the context and the expressed receptors on the target cells including the deleted in colorectal cancer (DCC), the uncoordinated-5 (UNC5), DSCAM, Neogenin-1, Adenosine A2b and Draxin receptors. By directing axonal growth cones toward the appropriate targets, netrin-1 is a critical actor in the formation of the intricate architecture of the nervous system. Netrin-1 is believed to be involved in additional biological and pathological processes in addition to its traditional function in neural development. The behavior of a diverse array of cell types is influenced by controlling cell adhesion and movement, which is impacted by netrin-1. It is a molecule of interest in both developmental biology and clinical research because of its involvement in angiogenesis, tumorigenesis, inflammation, and tissue regeneration, as confirmed by recent studies. The therapeutic capability of netrin-1 in disorders such as cancer, neurodegenerative disorders, and cardiovascular diseases warrants significant attention.
Collapse
Affiliation(s)
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bengaluru, Karnataka, India
| | - Mamata Chahar
- Department of Chemistry, NIMS University, Jaipur, Rajasthan, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | | | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | | | | | | |
Collapse
|
2
|
Nakamura Y, Miwa T, Shiga H, Sakata H, Shigeta D, Hatta T. Histological changes in the olfactory bulb and rostral migratory stream due to interruption of olfactory input. Auris Nasus Larynx 2024; 51:517-524. [PMID: 38522356 DOI: 10.1016/j.anl.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/14/2024] [Accepted: 01/23/2024] [Indexed: 03/26/2024]
Abstract
OBJECTIVE Periglomerular and granule cells in the adult mammalian olfactory bulb modulate olfactory signal transmission. These cells originate from the subventricular zone, migrate to the olfactory bulb via the Rostral Migratory Stream (RMS), and differentiate into mature cells within the olfactory bulb throughout postnatal life. While the regulation of neuroblast development is known to be affected by external stimuli, there is a lack of information concerning changes that occur during the recovery process after injury caused by external stimuli. To address this gap in research, the present study conducted histological observations to investigate changes in the olfactory bulb and RMS occurring after the degeneration and regeneration of olfactory neurons. METHODS To create a model of olfactory neurodegeneration, adult mice were administered methimazole intraperitoneally. Nasal tissue and whole brains were removed 3, 7, 14 and 28 days after methimazole administration, and EdU was administered 2 and 4 h before removal of these tissues to monitor dividing cells in the RMS. Methimazole-untreated mice were used as controls. Olfactory nerve fibers entering the olfactory glomerulus were observed immunohistochemically using anti-olfactory marker protein. In the brain tissue, the entire RMS was observed and the volume and total number of cells in the RMS were measured. In addition, the number of neuroblasts and dividing neuroblasts passing through the RMS were measured using anti-doublecortin and anti-EdU antibodies, respectively. Statistical analysis was performed using the Tukey test. RESULTS Olfactory epithelium degenerated was observed after methimazole administration, and recovered after 28 days. In the olfactory glomeruli, degeneration of OMP fibers began after methimazole administration, and after day 14, OMP fibers were reduced or absent by day 28, and overall OMP positive fibers were less than 20%. Glomerular volume tended to decrease after methimazole administration and did not appear to recover, even 28 days after recovery of the olfactory epithelium. In the RMS, EdU-positive cells decreased on day 3 and began to increase on day 7. However, they did not recover to the same levels as the control methimazole-untreated mice even after 28 days. CONCLUSION These results suggest that the division and maturation of neuroblasts migrating from the RMS was suppressed by olfactory nerve degeneration or the disruption of olfactory input.
Collapse
Affiliation(s)
- Yukari Nakamura
- Department of Otorhinolaryngology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Japan
| | - Takaki Miwa
- Department of Otorhinolaryngology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Japan.
| | - Hideaki Shiga
- Department of Otorhinolaryngology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Japan
| | - Hiromi Sakata
- Department of Anatomy I, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Japan
| | - Daichi Shigeta
- Department of Anatomy I, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Japan
| | - Toshihisa Hatta
- Department of Anatomy I, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Japan
| |
Collapse
|
3
|
Geribaldi-Doldán N, Carrascal L, Pérez-García P, Oliva-Montero JM, Pardillo-Díaz R, Domínguez-García S, Bernal-Utrera C, Gómez-Oliva R, Martínez-Ortega S, Verástegui C, Nunez-Abades P, Castro C. Migratory Response of Cells in Neurogenic Niches to Neuronal Death: The Onset of Harmonic Repair? Int J Mol Sci 2023; 24:6587. [PMID: 37047560 PMCID: PMC10095545 DOI: 10.3390/ijms24076587] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Harmonic mechanisms orchestrate neurogenesis in the healthy brain within specific neurogenic niches, which generate neurons from neural stem cells as a homeostatic mechanism. These newly generated neurons integrate into existing neuronal circuits to participate in different brain tasks. Despite the mechanisms that protect the mammalian brain, this organ is susceptible to many different types of damage that result in the loss of neuronal tissue and therefore in alterations in the functionality of the affected regions. Nevertheless, the mammalian brain has developed mechanisms to respond to these injuries, potentiating its capacity to generate new neurons from neural stem cells and altering the homeostatic processes that occur in neurogenic niches. These alterations may lead to the generation of new neurons within the damaged brain regions. Notwithstanding, the activation of these repair mechanisms, regeneration of neuronal tissue within brain injuries does not naturally occur. In this review, we discuss how the different neurogenic niches respond to different types of brain injuries, focusing on the capacity of the progenitors generated in these niches to migrate to the injured regions and activate repair mechanisms. We conclude that the search for pharmacological drugs that stimulate the migration of newly generated neurons to brain injuries may result in the development of therapies to repair the damaged brain tissue.
Collapse
Affiliation(s)
- Noelia Geribaldi-Doldán
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Livia Carrascal
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Patricia Pérez-García
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - José M. Oliva-Montero
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Ricardo Pardillo-Díaz
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Samuel Domínguez-García
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
- Department of Neuroscience, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Carlos Bernal-Utrera
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Fisioterapia, Facultad de Enfermería, Fisioterapia y Podología, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Ricardo Gómez-Oliva
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Sergio Martínez-Ortega
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| | - Cristina Verástegui
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Pedro Nunez-Abades
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Carmen Castro
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Departamento de Biomedicina, Biotecnología y Salud Pública, Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11003 Cádiz, Spain
| |
Collapse
|
4
|
Xie WS, Shehzadi K, Ma HL, Liang JH. A Potential Strategy for Treatment of Neurodegenerative Disorders by Regulation of Adult Hippocampal Neurogenesis in Human Brain. Curr Med Chem 2022; 29:5315-5347. [DOI: 10.2174/0929867329666220509114232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/13/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Adult hippocampal neurogenesis is a multistage mechanism that continues throughout the lifespan of human and non-human mammals. These adult-born neurons in the central nervous system (CNS) play a significant role in various hippocampus-dependent processes, including learning, mood regulation, pattern recognition, etc. Reduction of adult hippocampal neurogenesis, caused by multiple factors such as neurological disorders and aging, would impair neuronal proliferation and differentiation and result in memory loss. Accumulating studies have indicated that functional neuron impairment could be restored by promoting adult hippocampal neurogenesis. In this review, we summarized the small molecules that could efficiently promote the process of adult neurogenesis, particularly the agents that have the capacity of crossing the blood-brain barrier (BBB), and showed in vivo efficacy in mammalian brains. This may pave the way for the rational design of drugs to treat humnan neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Wei-Song Xie
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Kiran Shehzadi
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Hong-Le Ma
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Jian-Hua Liang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
- Yangtze Delta Region Academy of Beijing Institute of Technology, Jiaxing 314019, China
| |
Collapse
|
5
|
Schneider M, Vollmer L, Potthoff AL, Ravi VM, Evert BO, Rahman MA, Sarowar S, Kueckelhaus J, Will P, Zurhorst D, Joseph K, Maier JP, Neidert N, d’Errico P, Meyer-Luehmann M, Hofmann UG, Dolf A, Salomoni P, Güresir E, Enger PØ, Chekenya M, Pietsch T, Schuss P, Schnell O, Westhoff MA, Beck J, Vatter H, Waha A, Herrlinger U, Heiland DH. Meclofenamate causes loss of cellular tethering and decoupling of functional networks in glioblastoma. Neuro Oncol 2021; 23:1885-1897. [PMID: 33864086 PMCID: PMC8563322 DOI: 10.1093/neuonc/noab092] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Glioblastoma cells assemble to a syncytial communicating network based on tumor microtubes (TMs) as ultra-long membrane protrusions. The relationship between network architecture and transcriptional profile remains poorly investigated. Drugs that interfere with this syncytial connectivity such as meclofenamate (MFA) may be highly attractive for glioblastoma therapy. METHODS In a human neocortical slice model using glioblastoma cell populations of different transcriptional signatures, three-dimensional tumor networks were reconstructed, and TM-based intercellular connectivity was mapped on the basis of two-photon imaging data. MFA was used to modulate morphological and functional connectivity; downstream effects of MFA treatment were investigated by RNA sequencing and fluorescence-activated cell sorting (FACS) analysis. RESULTS TM-based network morphology strongly differed between the transcriptional cellular subtypes of glioblastoma and was dependent on axon guidance molecule expression. MFA revealed both a functional and morphological demolishment of glioblastoma network architectures which was reflected by a reduction of TM-mediated intercellular cytosolic traffic as well as a breakdown of TM length. RNA sequencing confirmed a downregulation of NCAM and axon guidance molecule signaling upon MFA treatment. Loss of glioblastoma communicating networks was accompanied by a failure in the upregulation of genes that are required for DNA repair in response to temozolomide (TMZ) treatment and culminated in profound treatment response to TMZ-mediated toxicity. CONCLUSION The capacity of TM formation reflects transcriptional cellular heterogeneity. MFA effectively demolishes functional and morphological TM-based syncytial network architectures. These findings might pave the way to a clinical implementation of MFA as a TM-targeted therapeutic approach.
Collapse
Affiliation(s)
- Matthias Schneider
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
- Brain Tumor Translational Research Affiliation, University Hospital Bonn, Bonn, Germany
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Lea Vollmer
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna-Laura Potthoff
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
- Brain Tumor Translational Research Affiliation, University Hospital Bonn, Bonn, Germany
| | - Vidhya M Ravi
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Neuroelectronic Systems, Medical Center, University of Freiburg, Freiburg, Germany
| | - Bernd O Evert
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | | | - Shahin Sarowar
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jan Kueckelhaus
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Paulina Will
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - David Zurhorst
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Kevin Joseph
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Neuroelectronic Systems, Medical Center, University of Freiburg, Freiburg, Germany
| | - Julian P Maier
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Nicolas Neidert
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Paolo d’Errico
- Department of Neurology, Medical Centre, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Centre, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrich G Hofmann
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Dolf
- Institute of Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Paolo Salomoni
- Nuclear Function in CNS Pathophysiology, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Per Ø Enger
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Torsten Pietsch
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Patrick Schuss
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
- Brain Tumor Translational Research Affiliation, University Hospital Bonn, Bonn, Germany
| | - Oliver Schnell
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jürgen Beck
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Andreas Waha
- Brain Tumor Translational Research Affiliation, University Hospital Bonn, Bonn, Germany
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Ulrich Herrlinger
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Dieter H Heiland
- Translational NeuroOncology Research Group, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Neuroelectronic Systems, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
Tucić M, Stamenković V, Andjus P. The Extracellular Matrix Glycoprotein Tenascin C and Adult Neurogenesis. Front Cell Dev Biol 2021; 9:674199. [PMID: 33996833 PMCID: PMC8117239 DOI: 10.3389/fcell.2021.674199] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Tenascin C (TnC) is a glycoprotein highly expressed in the extracellular matrix (ECM) during development and in the adult central nervous system (CNS) in regions of active neurogenesis, where neuron development is a tightly regulated process orchestrated by extracellular matrix components. In addition, newborn cells also communicate with glial cells, astrocytes and microglia, indicating the importance of signal integration in adult neurogenesis. Although TnC has been recognized as an important molecule in the regulation of cell proliferation and migration, complete regulatory pathways still need to be elucidated. In this review we discuss the formation of new neurons in the adult hippocampus and the olfactory system with specific reference to TnC and its regulating functions in this process. Better understanding of the ECM signaling in the niche of the CNS will have significant implications for regenerative therapies.
Collapse
Affiliation(s)
- Milena Tucić
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vera Stamenković
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle Andjus
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
7
|
Yao LL, Hu JX, Li Q, Lee D, Ren X, Zhang JS, Sun D, Zhang HS, Wang YG, Mei L, Xiong WC. Astrocytic neogenin/netrin-1 pathway promotes blood vessel homeostasis and function in mouse cortex. J Clin Invest 2021; 130:6490-6509. [PMID: 32853179 DOI: 10.1172/jci132372] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Astrocytes have multiple functions in the brain, including affecting blood vessel (BV) homeostasis and function. However, the underlying mechanisms remain elusive. Here, we provide evidence that astrocytic neogenin (NEO1), a member of deleted in colorectal cancer (DCC) family netrin receptors, is involved in blood vessel homeostasis and function. Mice with Neo1 depletion in astrocytes exhibited clustered astrocyte distribution and increased BVs in their cortices. These BVs were leaky, with reduced blood flow, disrupted vascular basement membranes (vBMs), decreased pericytes, impaired endothelial cell (EC) barrier, and elevated tip EC proliferation. Increased proliferation was also detected in cultured ECs exposed to the conditioned medium (CM) of NEO1-depleted astrocytes. Further screening for angiogenetic factors in the CM identified netrin-1 (NTN1), whose expression was decreased in NEO1-depleted cortical astrocytes. Adding NTN1 into the CM of NEO1-depleted astrocytes attenuated EC proliferation. Expressing NTN1 in NEO1 mutant cortical astrocytes ameliorated phenotypes in blood-brain barrier (BBB), EC, and astrocyte distribution. NTN1 depletion in astrocytes resulted in BV/BBB deficits in the cortex similar to those in Neo1 mutant mice. In aggregate, these results uncovered an unrecognized pathway, astrocytic NEO1 to NTN1, not only regulating astrocyte distribution, but also promoting cortical BV homeostasis and function.
Collapse
Affiliation(s)
- Ling-Ling Yao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jin-Xia Hu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Institute of Stroke Center and Department of Neurology, Xuzhou Medical University, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Qiang Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun-Shi Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neurology, Huaihe Hospital, Henan University, Kaifeng, Henan, China
| | - Dong Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hong-Sheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yong-Gang Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
8
|
Bressan C, Saghatelyan A. Intrinsic Mechanisms Regulating Neuronal Migration in the Postnatal Brain. Front Cell Neurosci 2021; 14:620379. [PMID: 33519385 PMCID: PMC7838331 DOI: 10.3389/fncel.2020.620379] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/08/2020] [Indexed: 01/19/2023] Open
Abstract
Neuronal migration is a fundamental brain development process that allows cells to move from their birthplaces to their sites of integration. Although neuronal migration largely ceases during embryonic and early postnatal development, neuroblasts continue to be produced and to migrate to a few regions of the adult brain such as the dentate gyrus and the subventricular zone (SVZ). In the SVZ, a large number of neuroblasts migrate into the olfactory bulb (OB) along the rostral migratory stream (RMS). Neuroblasts migrate in chains in a tightly organized micro-environment composed of astrocytes that ensheath the chains of neuroblasts and regulate their migration; the blood vessels that are used by neuroblasts as a physical scaffold and a source of molecular factors; and axons that modulate neuronal migration. In addition to diverse sets of extrinsic micro-environmental cues, long-distance neuronal migration involves a number of intrinsic mechanisms, including membrane and cytoskeleton remodeling, Ca2+ signaling, mitochondria dynamics, energy consumption, and autophagy. All these mechanisms are required to cope with the different micro-environment signals and maintain cellular homeostasis in order to sustain the proper dynamics of migrating neuroblasts and their faithful arrival in the target regions. Neuroblasts in the postnatal brain not only migrate into the OB but may also deviate from their normal path to migrate to a site of injury induced by a stroke or by certain neurodegenerative disorders. In this review, we will focus on the intrinsic mechanisms that regulate long-distance neuroblast migration in the adult brain and on how these pathways may be modulated to control the recruitment of neuroblasts to damaged/diseased brain areas.
Collapse
Affiliation(s)
- Cedric Bressan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| | - Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
9
|
Ribeiro FF, Xapelli S. An Overview of Adult Neurogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:77-94. [PMID: 34453294 DOI: 10.1007/978-3-030-74046-7_7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neurogenesis is maintained in the mammalian brain throughout adulthood in two main regions: the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the hippocampal dentate gyrus. Adult neurogenesis is a process composed of multiple steps by which neurons are generated from dividing adult neural stem cells and migrate to be integrated into existing neuronal circuits. Alterations in any of these steps impair neurogenesis and may compromise brain function, leading to cognitive impairment and neurodegenerative diseases. Therefore, understanding the cellular and molecular mechanisms that modulate adult neurogenesis is the centre of attention of regenerative research. In this chapter, we review the main properties of the adult neurogenic niches.
Collapse
Affiliation(s)
- Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
10
|
Jurkowski MP, Bettio L, K. Woo E, Patten A, Yau SY, Gil-Mohapel J. Beyond the Hippocampus and the SVZ: Adult Neurogenesis Throughout the Brain. Front Cell Neurosci 2020; 14:576444. [PMID: 33132848 PMCID: PMC7550688 DOI: 10.3389/fncel.2020.576444] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
Convincing evidence has repeatedly shown that new neurons are produced in the mammalian brain into adulthood. Adult neurogenesis has been best described in the hippocampus and the subventricular zone (SVZ), in which a series of distinct stages of neuronal development has been well characterized. However, more recently, new neurons have also been found in other brain regions of the adult mammalian brain, including the hypothalamus, striatum, substantia nigra, cortex, and amygdala. While some studies have suggested that these new neurons originate from endogenous stem cell pools located within these brain regions, others have shown the migration of neurons from the SVZ to these regions. Notably, it has been shown that the generation of new neurons in these brain regions is impacted by neurologic processes such as stroke/ischemia and neurodegenerative disorders. Furthermore, numerous factors such as neurotrophic support, pharmacologic interventions, environmental exposures, and stem cell therapy can modulate this endogenous process. While the presence and significance of adult neurogenesis in the human brain (and particularly outside of the classical neurogenic regions) is still an area of debate, this intrinsic neurogenic potential and its possible regulation through therapeutic measures present an exciting alternative for the treatment of several neurologic conditions. This review summarizes evidence in support of the classic and novel neurogenic zones present within the mammalian brain and discusses the functional significance of these new neurons as well as the factors that regulate their production. Finally, it also discusses the potential clinical applications of promoting neurogenesis outside of the classical neurogenic niches, particularly in the hypothalamus, cortex, striatum, substantia nigra, and amygdala.
Collapse
Affiliation(s)
- Michal P. Jurkowski
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Luis Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Emma K. Woo
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Anna Patten
- Centre for Interprofessional Clinical Simulation Learning (CICSL), Royal Jubilee Hospital, Victoria, BC, Canada
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Joana Gil-Mohapel
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
11
|
Louhivuori LM, Turunen PM, Louhivuori V, Al Rayyes I, Nordström T, Uhlén P, Åkerman KE. Neurotransmitters and Endothelins Acting on Radial Glial G-Protein-Coupled Receptors Are, Through Proteolytic NRG/ErbB4 Activation, Able to Modify the Migratory Behavior of Neocortical Cells and Mediate Bipolar-to-Multipolar Transition. Stem Cells Dev 2020; 29:1160-1177. [PMID: 31941419 DOI: 10.1089/scd.2019.0133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell-cell communication plays a central role in the guidance of migrating neurons during the development of the cerebral cortex. Neuregulins (NRGs) are essential mediators for migration and maintenance of the radial glial scaffold. We show, in this study that soluble NRG reduces neuronal motility, causes transition of bipolar cells to multipolar ones, and induces neuronal mitosis. Blocking the NRG receptor, ErbB4, results in reduction of neuron-neuron and neuron-radial glial contacts and causes an increase in neuronal motility. Blocking the radial glial metabotropic glutamate receptor 5 (mGluR5), the nonselective cation channel transient receptor potential 3 (TRPC3), or matrix metalloproteinases (MMPs) results in similar effects as ErbB4 blockade. Soluble NRG counteract the changes in motility pattern. Stimulation of other radial glial G-protein-coupled receptors (GPCRs), such as muscarinic acetylcholine receptors or endothelin receptors counteract all the effect of mGluR5 blockade, but not that of ErbB4, TRPC3, and MMP blockade. The results indicate that neurotransmitters and endothelins acting on radial glial GPCRs are, through proteolytic NRG/ErbB4 activation, able to modify the migratory behavior of neurons.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pauli M Turunen
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Verna Louhivuori
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Ibrahim Al Rayyes
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tommy Nordström
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karl E Åkerman
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Warren PM, Andrews MR, Smith M, Bartus K, Bradbury EJ, Verhaagen J, Fawcett JW, Kwok JCF. Secretion of a mammalian chondroitinase ABC aids glial integration at PNS/CNS boundaries. Sci Rep 2020; 10:11262. [PMID: 32647242 PMCID: PMC7347606 DOI: 10.1038/s41598-020-67526-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
Schwann cell grafts support axonal growth following spinal cord injury, but a boundary forms between the implanted cells and host astrocytes. Axons are reluctant to exit the graft tissue in large part due to the surrounding inhibitory environment containing chondroitin sulphate proteoglycans (CSPGs). We use a lentiviral chondroitinase ABC, capable of being secreted from mammalian cells (mChABC), to examine the repercussions of CSPG digestion upon Schwann cell behaviour in vitro. We show that mChABC transduced Schwann cells robustly secrete substantial quantities of the enzyme causing large-scale CSPG digestion, facilitating the migration and adhesion of Schwann cells on inhibitory aggrecan and astrocytic substrates. Importantly, we show that secretion of the engineered enzyme can aid the intermingling of cells at the Schwann cell-astrocyte boundary, enabling growth of neurites over the putative graft/host interface. These data were echoed in vivo. This study demonstrates the profound effect of the enzyme on cellular motility, growth and migration. This provides a cellular mechanism for mChABC induced functional and behavioural recovery shown in in vivo studies. Importantly, we provide in vitro evidence that mChABC gene therapy is equally or more effective at producing these effects as a one-time application of commercially available ChABC.
Collapse
Affiliation(s)
- Philippa M Warren
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK. .,Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, SE1 1UL, UK. .,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 0PY, UK.
| | - Melissa R Andrews
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK.,Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Marc Smith
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Katalin Bartus
- Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Elizabeth J Bradbury
- Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, SE1 1UL, UK
| | - Joost Verhaagen
- Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - James W Fawcett
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK.,Centre for Reconstructive Neuroscience, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Jessica C F Kwok
- Centre for Reconstructive Neuroscience, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
13
|
Matsushita K. Adhesion-stabilizing long-distance transport of cells on tissue surface. Phys Rev E 2020; 101:052410. [PMID: 32575308 DOI: 10.1103/physreve.101.052410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 05/12/2020] [Indexed: 11/07/2022]
Abstract
The stable transport of migrating eukaryotic cells is essential in organ development and repair processes. However, the mechanism that preserves transport stability over long distances in organs is not fully understood. As the driving mechanism of cell migration, the expressions of heterophilic cell-cell adhesion between moving cells and scaffolding tissue have been observed in such transport. In this paper, we theoretically investigate this heterophilic adhesion, which is persistently polarized in the migrating cell, as a possible transport stabilization mechanism. The adhesion was examined on the basis of the cellular Potts model, and our results confirm the stabilization of the transport to be an effect of the persistence.
Collapse
|
14
|
The use of bioactive matrices in regenerative therapies for traumatic brain injury. Acta Biomater 2020; 102:1-12. [PMID: 31751809 DOI: 10.1016/j.actbio.2019.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 01/01/2023]
Abstract
Functional deficits due to neuronal loss are a common theme across multiple neuropathologies, including traumatic brain injury (TBI). Apart from mitigating cell death, another approach to treating brain injuries involves re-establishing the neural circuitry at the lesion site by utilizing exogeneous and/or endogenous stem cells to achieve functional recovery. While there has been limited success, the emergence of new bioactive matrices that promote neural repair introduces new perspectives on the development of regenerative therapies for TBI. This review briefly discusses current development on cell-based therapies and the use of bioactive matrices, hydrogels in particular, when incorporated in regenerative therapies. Desirable characteristics of bioactive matrices that have been shown to augment neural repair in TBI models were identified and further discussed. Understanding the relative outcomes of newly developed biomaterials implanted in vivo can better guide the development of biomaterials as a therapeutic strategy, for biomaterial-based cellular therapies are still in their nascent stages. Nonetheless, the value of bioactive matrices as a treatment for acute brain injuries should be appreciated and further developed. STATEMENT OF SIGNIFICANCE: Cell-based therapies have received attention as an alternative therapeutic strategy to improve clinical outcome post-traumatic brain injury but have achieved limited success. Whilst the incorporation of newly developed biomaterials in regenerative therapies has shown promise in augmenting neural repair, studies have revealed new hurdles which must be overcome to improve their therapeutic efficacy. This review discusses the recent development of cell-based therapies with a specific focus on the use of bioactive matrices in the form of hydrogels, to complement cell transplantation within the injured brain. Moreover, this review consolidates in vivo animal studies that demonstrate relative functional outcome upon the implantation of different biomaterials to highlight their desirable traits to guide their development for regenerative therapies in traumatic brain injury.
Collapse
|
15
|
Fujioka T, Kaneko N, Sawamoto K. Blood vessels as a scaffold for neuronal migration. Neurochem Int 2019; 126:69-73. [PMID: 30851365 DOI: 10.1016/j.neuint.2019.03.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/19/2022]
Abstract
Neurogenesis and angiogenesis share regulatory factors that contribute to the formation of vascular networks and neuronal circuits in the brain. While crosstalk mechanisms between neural stem cells (NSCs) and the vasculature have been extensively investigated, recent studies have provided evidence that blood vessels also play an essential role in neuronal migration in the brain during development and regeneration. The mechanisms of the neuronal migration along blood vessels, referred to as "vascular-guided migration," are now being elucidated. The vascular endothelial cells secrete soluble factors that attract and promote neuronal migration in collaboration with astrocytes that enwrap the blood vessels. In addition, especially in the adult brain, the blood vessels serve as a migration scaffold for adult-born immature neurons generated in the ventricular-subventricular zone (V-SVZ), a germinal zone surrounding the lateral ventricles. The V-SVZ-derived immature neurons use the vascular scaffold to assist their migration toward an injured area after ischemic stroke, and contribute to neuronal regeneration. Here we review the current knowledge about the role of vasculature in neuronal migration and the molecular mechanisms controlling this process. While most of this research has been done in rodents, a comprehensive understanding of vasculature-guided neuronal migration could contribute to new therapeutic approaches for increasing new neurons in the brain after injury.
Collapse
Affiliation(s)
- Teppei Fujioka
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan; Department of Neurology and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan; Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8585, Japan.
| |
Collapse
|
16
|
Stem cell-secreted 14,15- epoxyeicosatrienoic acid rescues cholesterol homeostasis and autophagic flux in Niemann-Pick-type C disease. Exp Mol Med 2018; 50:1-14. [PMID: 30429460 PMCID: PMC6235958 DOI: 10.1038/s12276-018-0176-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/06/2018] [Accepted: 07/18/2018] [Indexed: 12/27/2022] Open
Abstract
We previously demonstrated that the direct transplantation of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) into the dentate gyrus ameliorated the neurological symptoms of Niemann–Pick type C1 (NPC1)-mutant mice. However, the clinical presentation of NPC1-mutant mice was not fully understood with a molecular mechanism. Here, we found 14,15-epoxyeicosatrienoic acid (14,15-EET), a cytochrome P450 (CYP) metabolite, from hUCB-MSCs and the cerebella of NPC1-mutant mice and investigated the functional consequence of this metabolite. Our screening of the CYP2J family indicated a dysregulation in the CYP system in a cerebellar-specific manner. Moreover, in Purkinje cells, CYP2J6 showed an elevated expression level compared to that of astrocytes, granule cells, and microglia. In this regard, we found that one CYP metabolite, 14,15-EET, acts as a key mediator in ameliorating cholesterol accumulation. In confirming this hypothesis, 14,15-EET treatment reduced the accumulation of cholesterol in human NPC1 patient-derived fibroblasts in vitro by suppressing cholesterol synthesis and ameliorating the impaired autophagic flux. We show that the reduced activity within the CYP system in the cerebellum could cause the neurological symptoms of NPC1 patients, as 14,15-EET treatment significantly rescued cholesterol accumulation and impaired autophagy. We also provide evidence that the intranasal administration of hUCB-MSCs is a highly promising alternative to traumatic surgical transplantation for NPC1 patients. An acid secreted by stem cells can reduce the excess cholesterol caused by a genetic metabolic disorder. Niemann–Pick type C disease is a rare, inherited condition that causes defective muscular development and progressive neurological degeneration. A key disease mechanism is the excessive accumulation of cholesterol within cells. Kyung-Sun Kang at Seoul National University, South Korea, and co-workers have demonstrated that a metabolite molecule called 14,15-epoxyeicosatrienoic acid (14,15-EET) derived from stem cells from human umbilical cord blood significantly reduced cholesterol in Neimann-Pick Type C mouse models and human cell samples. The team administered the stem cell therapy non-invasively via the nose, and observed significant improvements in motor function in the mice. Experiments in both animals and cells showed that the treatment resulted in reduced cholesterol levels and the correction of defective signalling within cells.
Collapse
|
17
|
Aoyagi Y, Hibi T, Kimori Y, Sawada M, Kawakami R, Sawamoto K, Nemoto T. Heterogeneous distribution of doublecortin-expressing cells surrounding the rostral migratory stream in the juvenile mouse. J Comp Neurol 2018; 526:2631-2646. [PMID: 30136724 DOI: 10.1002/cne.24521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/03/2018] [Accepted: 08/14/2018] [Indexed: 12/17/2022]
Abstract
In the postnatal mammalian brain, neural stem cells of the ventricular-subventricular zone continue to generate doublecortin (Dcx)-expressing immature neurons. Throughout life, these immature neurons migrate to the olfactory bulb through the rostral migratory stream (RMS). In this study, we investigated the distribution of these putative immature neurons using enhanced green fluorescent protein (EGFP) expression in the area surrounding the RMS of the juvenile Dcx-EGFP mice. Through the combined use of an optical clearing reagent (a 2,2'-thiodiethanol solution) and two-photon microscopy, we visualized three-dimensionally the EGFP-positive cells in the entire RMS and its surroundings. The resulting wide-field and high-definition images along with computational image processing methods developed in this study were used to comprehensively determine the position of the EGFP-positive cells. Our findings revealed that the EGFP-positive cells were heterogeneously distributed in the area surrounding the RMS. In addition, the orientation patterns of the leading process of these cells, which displayed the morphology of migrating immature neurons, differed depending on their location. These novel results provide highly precise morphological information for immature neurons and suggest that a portion of immature neurons may be detached from the RMS and migrate in various directions.
Collapse
Affiliation(s)
- Yuka Aoyagi
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Hokkaido, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Terumasa Hibi
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Hokkaido, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoshitaka Kimori
- Imaging Science Division, Center for Novel Science Initiatives, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Ryosuke Kawakami
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Hokkaido, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan.,Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Tomomi Nemoto
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Hokkaido, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
18
|
Hsu WC, Yu CH, Kung WM, Huang KF. Enhancement of matrix metalloproteinases 2 and 9 accompanied with neurogenesis following collagen glycosaminoglycan matrix implantation after surgical brain injury. Neural Regen Res 2018; 13:1007-1012. [PMID: 29926827 PMCID: PMC6022476 DOI: 10.4103/1673-5374.233443] [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] [Accepted: 04/02/2018] [Indexed: 02/05/2023] Open
Abstract
Surgical brain injury may result in irreversible neurological deficits. Our previous report showed that partial regeneration of a traumatic brain lesion is achieved by implantation of collagen glycosaminoglycan (CGM). Matrix metalloproteinases (MMPs) may play an important role in neurogenesis but there is currently a lack of studies displaying the relationship between the stimulation of MMPs and neurogenesis after collagen glycosaminoglycan implantation following surgical brain trauma. The present study was carried out to further examine the expression of MMP2 and MMP9 after implantation of collagen glycosaminoglycan (CGM) following surgical brain trauma. Using the animal model of surgically induced brain lesion, we implanted CGM into the surgical trauma. Rats were thus divided into three groups: (1) sham operation group: craniotomy only; (2) lesion (L) group: craniotomy + surgical trauma lesion; (3) lesion + CGM (L + CGM) group: CGM implanted following craniotomy and surgical trauma lesion. Cells positive for SOX2 (marker of proliferating neural progenitor cells) and matrix metalloproteinases (MMP2 and MMP9) in the lesion boundary zone were assayed and analyzed by immunofluorescence and ELISA commercial kits, respectively. Our results demonstrated that following implantation of CGM after surgical brain trauma, significant increases in MMP2+/SOX2+ cells and MMP9+/SOX2+ cells were seen within the lesion boundary zone in the L + CGM group. Tissue protein concentrations of MMP2 and MMP9 also increased after CGM scaffold implantation. These findings suggest that implantation of a CGM scaffold alone after surgical brain trauma can enhance the expression of MMP2 and MMP9 accompanied by neurogenesis.
Collapse
Affiliation(s)
- Wei-Cherng Hsu
- Department of Ophthalmology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan, China
| | - Chun-Hsien Yu
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan, China
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
| | - Woon-Man Kung
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
- Department of Exercise and Health Promotion, College of Education, Chinese Culture University, Taipei, Taiwan, China
| | - Kuo-Feng Huang
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan, China
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
| |
Collapse
|
19
|
Andreopoulou E, Arampatzis A, Patsoni M, Kazanis I. Being a Neural Stem Cell: A Matter of Character But Defined by the Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:81-118. [PMID: 29204830 DOI: 10.1007/978-3-319-69194-7_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cells that build the nervous system, either this is a small network of ganglia or a complicated primate brain, are called neural stem and progenitor cells. Even though the very primitive and the very recent neural stem cells (NSCs) share common basic characteristics that are hard-wired within their character, such as the expression of transcription factors of the SoxB family, their capacity to give rise to extremely different neural tissues depends significantly on instructions from the microenvironment. In this chapter we explore the nature of the NSC microenvironment, looking through evolution, embryonic development, maturity and even disease. Experimental work undertaken over the last 20 years has revealed exciting insight into the NSC microcosmos. NSCs are very capable in producing their own extracellular matrix and in regulating their behaviour in an autocrine and paracrine manner. Nevertheless, accumulating evidence indicates an important role for the vasculature, especially within the NSC niches of the postnatal brain; while novel results reveal direct links between the metabolic state of the organism and the function of NSCs.
Collapse
Affiliation(s)
- Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Asterios Arampatzis
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Melina Patsoni
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Ilias Kazanis
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece.
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Turunen PM, Louhivuori LM, Louhivuori V, Kukkonen JP, Åkerman KE. Endocannabinoid Signaling in Embryonic Neuronal Motility and Cell–Cell Contact – Role of mGluR5 and TRPC3 Channels. Neuroscience 2018; 375:135-148. [DOI: 10.1016/j.neuroscience.2018.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 01/31/2018] [Accepted: 02/02/2018] [Indexed: 10/18/2022]
|
21
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
22
|
|
23
|
Belvindrah R, Natarajan K, Shabajee P, Bruel-Jungerman E, Bernard J, Goutierre M, Moutkine I, Jaglin XH, Savariradjane M, Irinopoulou T, Poncer JC, Janke C, Francis F. Mutation of the α-tubulin Tuba1a leads to straighter microtubules and perturbs neuronal migration. J Cell Biol 2017; 216:2443-2461. [PMID: 28687665 PMCID: PMC5551700 DOI: 10.1083/jcb.201607074] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 05/05/2017] [Accepted: 06/01/2017] [Indexed: 12/24/2022] Open
Abstract
Mutation of α-tubulin isotypes is associated with cortical malformations. Belvindrah et al. show that Tuba1 mutation leads to impaired neuronal saltatory migration in vivo as a result of functional and structural microtubule defects. Comparative analyses of Tuba1a and Tuba8 in tubulin heterodimer structure and microtubule polymerization reveal an essential, noncompensated role for Tuba1a in the neuronal rostral migratory system. Brain development involves extensive migration of neurons. Microtubules (MTs) are key cellular effectors of neuronal displacement that are assembled from α/β-tubulin heterodimers. Mutation of the α-tubulin isotype TUBA1A is associated with cortical malformations in humans. In this study, we provide detailed in vivo and in vitro analyses of Tuba1a mutants. In mice carrying a Tuba1a missense mutation (S140G), neurons accumulate, and glial cells are dispersed along the rostral migratory stream in postnatal and adult brains. Live imaging of Tuba1a-mutant neurons revealed slowed migration and increased neuronal branching, which correlated with directionality alterations and perturbed nucleus–centrosome (N–C) coupling. Tuba1a mutation led to increased straightness of newly polymerized MTs, and structural modeling data suggest a conformational change in the α/β-tubulin heterodimer. We show that Tuba8, another α-tubulin isotype previously associated with cortical malformations, has altered function compared with Tuba1a. Our work shows that Tuba1a plays an essential, noncompensated role in neuronal saltatory migration in vivo and highlights the importance of MT flexibility in N–C coupling and neuronal-branching regulation during neuronal migration.
Collapse
Affiliation(s)
- Richard Belvindrah
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Kathiresan Natarajan
- Institut Curie, Paris Sciences et Lettres Research Université (PSL), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS), UMR 3348, Orsay, France
| | - Preety Shabajee
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Elodie Bruel-Jungerman
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Jennifer Bernard
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Marie Goutierre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Imane Moutkine
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Xavier H Jaglin
- Department of Neuroscience and Physiology, Smilow Neuroscience Program, Neuroscience Institute, New York University, New York, NY
| | - Mythili Savariradjane
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Theano Irinopoulou
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Jean-Christophe Poncer
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Carsten Janke
- Institut Curie, Paris Sciences et Lettres Research Université (PSL), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS), UMR 3348, Orsay, France
| | - Fiona Francis
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR S-839, Paris, France .,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Université Paris 06, UMR S-839, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
24
|
Miyakoshi LM, Marques-Coelho D, De Souza LER, Lima FRS, Martins VR, Zanata SM, Hedin-Pereira C. Evidence of a Cell Surface Role for Hsp90 Complex Proteins Mediating Neuroblast Migration in the Subventricular Zone. Front Cell Neurosci 2017; 11:138. [PMID: 28567003 PMCID: PMC5434112 DOI: 10.3389/fncel.2017.00138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/26/2017] [Indexed: 11/30/2022] Open
Abstract
In most mammalian brains, the subventricular zone (SVZ) is a germinative layer that maintains neurogenic activity throughout adulthood. Neuronal precursors arising from this region migrate through the rostral migratory stream (RMS) and reach the olfactory bulbs where they differentiate and integrate into the local circuitry. Recently, studies have shown that heat shock proteins have an important role in cancer cell migration and blocking Hsp90 function was shown to hinder cell migration in the developing cerebellum. In this work, we hypothesize that chaperone complexes may have an important function regulating migration of neuronal precursors from the subventricular zone. Proteins from the Hsp90 complex are present in the postnatal SVZ as well as in the RMS. Using an in vitro SVZ explant model, we have demonstrated the expression of Hsp90 and Hop/STI1 by migrating neuroblasts. Treatment with antibodies against Hsp90 and co-chaperone Hop/STI1, as well as Hsp90 and Hsp70 inhibitors hinder neuroblast chain migration. Time-lapse videomicroscopy analysis revealed that cell motility and average migratory speed was decreased after exposure to both antibodies and inhibitors. Antibodies recognizing Hsp90, Hsp70, and Hop/STI1 were found bound to the membranes of cells from primary SVZ cultures and biotinylation assays demonstrated that Hsp70 and Hop/STI1 could be found on the external leaflet of neuroblast membranes. The latter could also be detected in conditioned medium samples obtained from cultivated SVZ cells. Our results suggest that chaperones Hsp90, Hsp70, and co-chaperone Hop/STI1, components of the Hsp90 complex, regulate SVZ neuroblast migration in a concerted manner through an extracellular mechanism.
Collapse
Affiliation(s)
- Leo M Miyakoshi
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Laboratory of Cellular NeuroAnatomy, Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Diego Marques-Coelho
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Laboratory of Cellular NeuroAnatomy, Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Luiz E R De Souza
- Department of Basic Pathology, Federal University of ParanáParaná, Brazil
| | - Flavia R S Lima
- Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Vilma R Martins
- International Research Center, A.C. Camargo Cancer CenterSão Paulo, Brazil
| | - Silvio M Zanata
- Department of Basic Pathology, Federal University of ParanáParaná, Brazil
| | - Cecilia Hedin-Pereira
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Laboratory of Cellular NeuroAnatomy, Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,VPPLR-Fundação Oswaldo Cruz (Fiocruz)Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Kaneko N, Sawada M, Sawamoto K. Mechanisms of neuronal migration in the adult brain. J Neurochem 2017; 141:835-847. [DOI: 10.1111/jnc.14002] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/06/2017] [Accepted: 02/21/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Naoko Kaneko
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medial Sciences; Nagoya Aichi Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medial Sciences; Nagoya Aichi Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medial Sciences; Nagoya Aichi Japan
- Division of Neural Development and Regeneration; National Institute for Physiological Sciences; Okazaki Aichi Japan
| |
Collapse
|
26
|
|
27
|
Lu H, Song X, Wang F, Wang G, Wu Y, Wang Q, Wang Y, Yang GY, Zhang Z. Hyperexpressed Netrin-1 Promoted Neural Stem Cells Migration in Mice after Focal Cerebral Ischemia. Front Cell Neurosci 2016; 10:223. [PMID: 27746720 PMCID: PMC5042963 DOI: 10.3389/fncel.2016.00223] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/16/2016] [Indexed: 11/13/2022] Open
Abstract
Endogenous Netrin-1 (NT-1) protein was significantly increased after cerebral ischemia, which may participate in the repair after transient cerebral ischemic injury. In this work, we explored whether NT-1 can be steadily overexpressed by adeno-associated virus (AAV) and the exogenous NT-1 can promote neural stem cells migration from the subventricular zone (SVZ) region after cerebral ischemia. Adult CD-1 mice were injected stereotacticly with AAV carrying NT-1 gene (AAV-NT-1). Mice underwent 60 min of middle cerebral artery (MCA) occlusion 1 week after injection. We found that NT-1 mainly expressed in neuron and astrocyte, and the expression level of NT-1 significantly increased 1 week after AAV-NT-1 gene transfer and lasted for 28 days, even after transient middle cerebral artery occlusion (tMCAO) as well (p < 0.05). Immunohistochemistry results showed that the number of neural stem cells was greatly increased in the SVZ region of AAV-NT-1-transduced mice compared with control mice. Our study showed that overexpressed NT-1 promoted neural stem cells migration from SVZ. This result suggested that NT-1 is a promising factor for repairing and remodeling after focal cerebral ischemia.
Collapse
Affiliation(s)
- Haiyan Lu
- Department of Neurology, Shanghai General Hospital, Shanghai JiaoTong University Shanghai, China
| | - Xiaoyan Song
- Department of Neurology, Shanghai General Hospital, Shanghai JiaoTong University Shanghai, China
| | - Feng Wang
- Department of Neurology, Shanghai General Hospital, Shanghai JiaoTong University Shanghai, China
| | - Guodong Wang
- Department of Neurology, Shanghai General Hospital, Shanghai JiaoTong University Shanghai, China
| | - Yuncheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai JiaoTong University Shanghai, China
| | - Qiaoshu Wang
- Department of Neurology, Shanghai General Hospital, Shanghai JiaoTong University Shanghai, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai Jiao Tong University Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai Jiao Tong University Shanghai, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|
28
|
Huilgol D, Tole S. Cell migration in the developing rodent olfactory system. Cell Mol Life Sci 2016; 73:2467-90. [PMID: 26994098 PMCID: PMC4894936 DOI: 10.1007/s00018-016-2172-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 02/08/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
The components of the nervous system are assembled in development by the process of cell migration. Although the principles of cell migration are conserved throughout the brain, different subsystems may predominantly utilize specific migratory mechanisms, or may display unusual features during migration. Examining these subsystems offers not only the potential for insights into the development of the system, but may also help in understanding disorders arising from aberrant cell migration. The olfactory system is an ancient sensory circuit that is essential for the survival and reproduction of a species. The organization of this circuit displays many evolutionarily conserved features in vertebrates, including molecular mechanisms and complex migratory pathways. In this review, we describe the elaborate migrations that populate each component of the olfactory system in rodents and compare them with those described in the well-studied neocortex. Understanding how the components of the olfactory system are assembled will not only shed light on the etiology of olfactory and sexual disorders, but will also offer insights into how conserved migratory mechanisms may have shaped the evolution of the brain.
Collapse
Affiliation(s)
- Dhananjay Huilgol
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Cold Spring Harbor Laboratory, Cold Spring Harbor, USA
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
29
|
Low Density Lipoprotein Receptor Related Proteins as Regulators of Neural Stem and Progenitor Cell Function. Stem Cells Int 2016; 2016:2108495. [PMID: 26949399 PMCID: PMC4754494 DOI: 10.1155/2016/2108495] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/24/2015] [Accepted: 01/06/2016] [Indexed: 12/20/2022] Open
Abstract
The central nervous system (CNS) is a highly organised structure. Many signalling systems work in concert to ensure that neural stem cells are appropriately directed to generate progenitor cells, which in turn mature into functional cell types including projection neurons, interneurons, astrocytes, and oligodendrocytes. Herein we explore the role of the low density lipoprotein (LDL) receptor family, in particular family members LRP1 and LRP2, in regulating the behaviour of neural stem and progenitor cells during development and adulthood. The ability of LRP1 and LRP2 to bind a diverse and extensive range of ligands, regulate ligand endocytosis, recruit nonreceptor tyrosine kinases for direct signal transduction and signal in conjunction with other receptors, enables them to modulate many crucial neural cell functions.
Collapse
|
30
|
Tsupykov O, Kanemitsu M, Smozhanik E, Skibo G, Dayer AG, Kiss JZ. Relationship of Grafted FGF-2-Overexpressing Neural Stem/Progenitor Cells With the Vasculature in the Cerebral Cortex. Cell Transplant 2016; 25:1359-69. [PMID: 26810970 DOI: 10.3727/096368916x690421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neural progenitor cells (NPCs) overexpressing fibroblast growth factor 2 (FGF-2) have the distinct tendency to associate with the vasculature and establish multiple proliferative clusters in the perivascular environment after transplantation into the cerebral cortex. Strikingly, the vascular clusters of progenitor cells give rise to immature neurons after ischemic injury, raising prospects for the formation of ectopic neurogenic niches for repair. We investigated the spatial relationship of perivascular clusters with the host vascular structures. FGF-2-GFP-transduced NPCs were transplanted into the intact somatosensory rat cortex. Confocal microscopic analysis revealed that grafted cells preferentially contacted venules at sites with aquaporin-4-positive astrocytic endfeet and avoided contacts with desmin-positive pericytes. Electron microscopic analysis confirmed that grafted cells preferentially made contact with astroglial endfeet, and only a minority of them reached the endothelial basal lamina. These results provide new insights into the fine structural and anatomical relationship between grafted FGF-2-transduced NPCs and the host vasculature.
Collapse
Affiliation(s)
- Oleg Tsupykov
- Department of Cytology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | | | | | | | | | | |
Collapse
|
31
|
Röckle I, Hildebrandt H. Deficits of olfactory interneurons in polysialyltransferase- and NCAM-deficient mice. Dev Neurobiol 2015; 76:421-33. [PMID: 26153130 DOI: 10.1002/dneu.22324] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 07/03/2015] [Accepted: 07/03/2015] [Indexed: 11/09/2022]
Abstract
The neurogenic niche of the anterior subventricular zone (SVZ) persistently generates neuroblasts, which migrate along the rostral migratory stream (RMS) into the olfactory bulb (OB), where they differentiate into granule and periglomerular cells. Loss of the neural cell adhesion molecule NCAM or its post-translational modification polysialic acid (polySia) impairs migration causing accumulations of cells in the proximal RMS and decreased OB volume. Polysialylation of NCAM is implemented by two polysialyltransferases, ST8SIA2 and ST8SIA4, with overlapping functions. Here, we used mice with Ncam1 and polysialyltransferase deletions to analyze how partial or complete loss of polySia synthesis or a combined loss of polySia and NCAM affects the RMS and the interneuron composition in the OB. Numerous calretinin (CR)-positive cells were detected dispersed around the RMS in Ncam1 knockout, St8sia2, St8sia4 double-knockout, and St8sia2, St8sia4, Ncam1 triple-knockout mice, as well as in St8sia2(-/-) but not in St8sia4(-/-) mice. These changes were not reflected by reductions of CR-positive cells in the granule or glomerular layer of the OB. Instead, calbindin-positive periglomerular interneurons were strongly reduced in all polySia-NCAM negative mice and slightly attenuated in St8sia2(-/-) as well as in the St8sia4(-/-) mice, which were devoid of ectopic CR-positive cells along the RMS. Consistent with the early developmental generation of calbindin- as compared with CR-positive OB interneurons, this phenotype was fully developed at postnatal day 5. Together, these results demonstrate that the early development of calbindin-positive periglomerular interneurons depends on the presentation of polySia on NCAM and requires the activity of both polysialyltransferases.
Collapse
Affiliation(s)
- Iris Röckle
- Institute of Cellular Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Herbert Hildebrandt
- Institute of Cellular Chemistry, Hannover Medical School, Hannover, 30625, Germany.,Center for Systems Neuroscience Hannover (ZSN), Hannover, Germany
| |
Collapse
|
32
|
Bakhshetyan K, Saghatelyan A. Tracking Neuronal Migration in Adult Brain Slices. ACTA ACUST UNITED AC 2015; 71:3.28.1-3.28.13. [DOI: 10.1002/0471142301.ns0328s71] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Karen Bakhshetyan
- Cellular Neurobiology Unit, Institut Universitaire en santé mentale de Québec Quebec City Canada
| | - Armen Saghatelyan
- Cellular Neurobiology Unit, Institut Universitaire en santé mentale de Québec Quebec City Canada
- Department of Psychiatry and Neuroscience, Université Laval Quebec City Canada
| |
Collapse
|
33
|
Xu CJ, Wang JL, Jin WL. The Neural Stem Cell Microenvironment: Focusing on Axon Guidance Molecules and Myelin-Associated Factors. J Mol Neurosci 2015; 56:887-897. [PMID: 25757451 DOI: 10.1007/s12031-015-0538-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/27/2015] [Indexed: 12/20/2022]
Abstract
Neural stem cells (NSCs) could produce various cell phenotypes in the subventricular zone (SVZ) and dentate gyrus of the hippocampus in the central nervous system (CNS), where neurogenesis has been determined to occur. The extracellular microenvironment also influences the behaviors of NSCs during development and at CNS injury sites. Our previous study indicates that myelin, a component of the CNS, could regulate the differentiation of NSCs in vitro. Recent reports have implicated three myelin-derived inhibitors, NogoA, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp), as well as several axon guidance molecules as regulators of NSC survival, proliferation, migration, and differentiation. However, the molecular mechanisms underlying the behavior of NSCs are not fully understood. In this study, we summarize the current literature on the effects of different extrinsic factors on NSCs and discuss possible mechanisms, as well as future possible clinical applications.
Collapse
Affiliation(s)
- Chao-Jin Xu
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, University town, Cha Shan, Zhejiang, 325035, China.
| | - Jun-Ling Wang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China. .,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, 200240, China.
| |
Collapse
|
34
|
Regulation of subventricular zone-derived cells migration in the adult brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 853:1-21. [PMID: 25895704 DOI: 10.1007/978-3-319-16537-0_1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The subventricular zone of the lateral ventricles (SVZ) is the largest source of neural stem cells (NSCs) in the adult mammalian brain. Newly generated neuroblasts from the SVZ form cellular chains that migrate through the rostral migratory stream (RMS) into the olfactory bulb (OB), where they become mature neurons. Migration through the RMS is a highly regulated process of intrinsic and extrinsic factors, orchestrated to achieve direction and integration of neuroblasts into OB circuitry. These factors include internal cytoskeletal and volume regulators, extracellular matrix proteins, and chemoattractant and chemorepellent proteins. All these molecules direct the cells away from the SVZ, through the RMS, and into the OB guaranteeing their correct integration. Following brain injury, some neuroblasts escape the RMS and migrate into the lesion site to participate in regeneration, a phenomenon that is also observed with brain tumors. This review focuses on factors that regulate the migration of SVZ precursor cells in the healthy and pathologic brain. A better understanding of the factors that control the movement of newly generated cells may be crucial for improving the use of NSC-replacement therapy for specific neurological diseases.
Collapse
|
35
|
Louhivuori LM, Jansson L, Turunen PM, Jäntti MH, Nordström T, Louhivuori V, Åkerman KE. Transient receptor potential channels and their role in modulating radial glial-neuronal interaction: a signaling pathway involving mGluR5. Stem Cells Dev 2014; 24:701-13. [PMID: 25347706 DOI: 10.1089/scd.2014.0209] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The guidance of developing neurons to the right position in the central nervous system is of central importance in brain development. Canonical transient receptor potential (TRPC) channels are thought to mediate turning responses of growth cones to guidance cues through fine control of calcium transients. Proliferating and 1- to 5-day-differentiated neural progenitor cells (NPCs) showed expression of Trpc1 and Trpc3 mRNA, while Trpc4-7 was not clearly detected. Time-lapse imaging showed that the motility pattern of neuronal cells was phasic with bursts of rapid movement (>60 μm/h), changes in direction, and intermittent slow phases or stallings (<40 μm/h), which frequently occurred in close contact with radial glial processes. Genetic interference with the TRPC3 and TRPC1 channel enhanced the motility of NPCs (burst frequency/stalling frequency). TRPC3-deficient cells or cells treated with the TRPC3 blocker pyr3 infrequently changed direction and seldom contacted radial glial processes. TRPC channels are also activated by group I metabotropic glutamate receptors (mGluR1 and mGluR5). As shown here, pyr3 blocked the calcium response mediated through mGluR5 in radial glial processes. Furthermore, 2-methyl-6-(phenylethynyl)pyridine, a blocker of mGluR5, affected the motility pattern in a similar way as TRPC3/6 double knockout or pyr3. The results suggest that radial glial cells exert attractant signals to migrating neuronal cells, which alter their motility pattern. Our results suggest that mGluR5 acting through TRPC3 is of central importance in radial glial-mediated neuronal guidance.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- Biomedicum Helsinki, Institute of Biomedicine/Physiology, University of Helsinki , Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
36
|
Cuccioli V, Bueno C, Belvindrah R, Lledo PM, Martinez S. Attractive action of FGF-signaling contributes to the postnatal developing hippocampus. Hippocampus 2014; 25:486-99. [DOI: 10.1002/hipo.22386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Affiliation(s)
- V. Cuccioli
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández (CSIC-UMH); Sant Joan d'Alacant 03550 Spain
| | - C. Bueno
- Faculty of Medicine; Department of Human Anatomy and Psychobiology; IMIB-Arrixaca and CIBERSAM (Centro Investigación Biomedica en Red Salud Mental), University of Murcia E-30071; Murcia Spain
| | - R. Belvindrah
- Laboratory for Perception and Memory; Institut Pasteur; F-75015 Paris France
- Centre National de la Recherche Scientifique (CNRS); Unité Mixte de Recherche 3571 F-75015 Paris France
| | - P.-M. Lledo
- Laboratory for Perception and Memory; Institut Pasteur; F-75015 Paris France
- Centre National de la Recherche Scientifique (CNRS); Unité Mixte de Recherche 3571 F-75015 Paris France
| | - S. Martinez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández (CSIC-UMH); Sant Joan d'Alacant 03550 Spain
- Faculty of Medicine; Department of Human Anatomy and Psychobiology; IMIB-Arrixaca and CIBERSAM (Centro Investigación Biomedica en Red Salud Mental), University of Murcia E-30071; Murcia Spain
| |
Collapse
|
37
|
Donega V, Nijboer CH, van Tilborg G, Dijkhuizen RM, Kavelaars A, Heijnen CJ. Intranasally administered mesenchymal stem cells promote a regenerative niche for repair of neonatal ischemic brain injury. Exp Neurol 2014; 261:53-64. [PMID: 24945601 DOI: 10.1016/j.expneurol.2014.06.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/30/2014] [Accepted: 06/09/2014] [Indexed: 12/15/2022]
Abstract
Previous work from our group has shown that intranasal MSC-treatment decreases lesion volume and improves motor and cognitive behavior after hypoxic-ischemic (HI) brain damage in neonatal mice. Our aim was to determine the kinetics of MSC migration after intranasal administration, and the early effects of MSCs on neurogenic processes and gliosis at the lesion site. HI brain injury was induced in 9-day-old mice and MSCs were administered intranasally at 10days post-HI. The kinetics of MSC migration were investigated by immunofluorescence and MRI analysis. BDNF and NGF gene expression was determined by qPCR analysis following MSC co-culture with HI brain extract. Nestin, Doublecortin, NeuN, GFAP, Iba-1 and M1/M2 phenotypic expression was assessed over time. MRI and immunohistochemistry analyses showed that MSCs reach the lesion site already within 2h after intranasal administration. At 12h after administration the number of MSCs at the lesion site peaks and decreases significantly at 72h. The number of DCX(+) cells increased 1 to 3days after MSC administration in the SVZ. At the lesion, GFAP(+)/nestin(+) and DCX(+) expression increased 3 to 5days after MSC-treatment. The number of NeuN(+) cells increased within 5days, leading to a dramatic regeneration of the somatosensory cortex and hippocampus at 18days after intranasal MSC administration. Interestingly, MSCs expressed significantly more BDNF gene when exposed to HI brain extract in vitro. Furthermore, MSC-treatment resulted in the resolution of the glial scar surrounding the lesion, represented by a decrease in reactive astrocytes and microglia and polarization of microglia towards the M2 phenotype. In view of the current lack of therapeutic strategies, we propose that intranasal MSC administration is a powerful therapeutic option through its functional repair of the lesion represented by regeneration of the cortical and hippocampal structure and decrease of gliosis.
Collapse
Affiliation(s)
- Vanessa Donega
- Lab. of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cora H Nijboer
- Lab. of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Geralda van Tilborg
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, The Netherlands
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, The Netherlands
| | - Annemieke Kavelaars
- Lab. of Neuroimmunology, Department of Symptom Research, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Cobi J Heijnen
- Lab. of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands; Lab. of Neuroimmunology, Department of Symptom Research, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
38
|
|
39
|
Das A, Gajendra S, Falenta K, Oudin MJ, Peschard P, Feng S, Wu B, Marshall CJ, Doherty P, Guo W, Lalli G. RalA promotes a direct exocyst-Par6 interaction to regulate polarity in neuronal development. J Cell Sci 2014; 127:686-99. [PMID: 24284074 PMCID: PMC4007768 DOI: 10.1242/jcs.145037] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/05/2013] [Indexed: 01/25/2023] Open
Abstract
Cell polarization is essential for neuronal development in both the embryonic and postnatal brain. Here, using primary cultures, in vivo postnatal electroporation and conditional genetic ablation, we show that the Ras-like small GTPase RalA and its effector, the exocyst, regulate the morphology and polarized migration of neural progenitors derived from the subventricular zone, a major neurogenic niche in the postnatal brain. Active RalA promotes the direct binding between the exocyst subunit Exo84 and the PDZ domain of Par6 through a non-canonical PDZ-binding motif. Blocking the Exo84-Par6 interaction impairs polarization in postnatal neural progenitors and cultured embryonic neurons. Our results provide the first in vivo characterization of RalA function in the mammalian brain and highlight a novel molecular mechanism for cell polarization. Given that the exocyst and the Par complex are conserved in many tissues, the functional significance of their interaction and its regulation by RalA are likely to be important in a wide range of polarization events.
Collapse
Affiliation(s)
- Amlan Das
- University of Pennsylvania Department of Biology, Philadelphia, PA 19104, USA
| | - Sangeetha Gajendra
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Katarzyna Falenta
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Madeleine J. Oudin
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Pascal Peschard
- The Institute of Cancer Research, Division of Cancer Cell Biology, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK
| | - Shanshan Feng
- University of Pennsylvania Department of Biology, Philadelphia, PA 19104, USA
| | - Bin Wu
- University of Pennsylvania Department of Biology, Philadelphia, PA 19104, USA
| | - Christopher J. Marshall
- The Institute of Cancer Research, Division of Cancer Cell Biology, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK
| | - Patrick Doherty
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Wei Guo
- University of Pennsylvania Department of Biology, Philadelphia, PA 19104, USA
| | - Giovanna Lalli
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
40
|
Fon D, Zhou K, Ercole F, Fehr F, Marchesan S, Minter MR, Crack PJ, Finkelstein DI, Forsythe JS. Nanofibrous scaffolds releasing a small molecule BDNF-mimetic for the re-direction of endogenous neuroblast migration in the brain. Biomaterials 2014; 35:2692-712. [PMID: 24406218 DOI: 10.1016/j.biomaterials.2013.12.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/10/2013] [Indexed: 01/19/2023]
Abstract
Brain tissue engineering has the potential to harness existing elements of neurogenesis within the adult brain to overcome a microenvironment that is otherwise inhibitory to regeneration, especially following severe tissue damage. This study investigates the ability of electrospun poly ε-caprolactone (PCL) to re-direct the migratory pathway of endogenous neuroblasts from the disrupted subventricular zone (SVZ). A small molecule non-peptide ligand (BDNF-mimetic) that mimicked the trophic properties of brain-derived neurotrophic factor (BDNF) was incorporated into electrospun PCL scaffolds to improve neuroblast survival and promote neuroblast migration towards the implant. PCL scaffolds were able to support neuroblast infiltration and migration along the implant tract. In the presence of the BDNF-mimetic, neuroblasts were able to migrate towards the implant via the parenchyma, and their persistence within the implants was prolonged. In addition, the BDNF-mimetic improved implant integration and increased local neuronal plasticity by increasing neurite sprouting at the tissue-implant interface. SMI32+ neurites were observed inside scaffolds at 21 days but not 8 days post implantation, indicating that at least some of the infiltrated neuroblasts had differentiated into neurons.
Collapse
Affiliation(s)
- Deniece Fon
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia; Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kun Zhou
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Francesca Ercole
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Friederike Fehr
- CSIRO Materials Science and Engineering, Clayton, VIC 3053, Australia
| | - Silvia Marchesan
- CSIRO Materials Science and Engineering, Clayton, VIC 3053, Australia
| | - Myles R Minter
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David I Finkelstein
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - John S Forsythe
- Department of Materials Engineering, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
41
|
Hikita T, Ohno A, Sawada M, Ota H, Sawamoto K. Rac1-mediated indentation of resting neurons promotes the chain migration of new neurons in the rostral migratory stream of post-natal mouse brain. J Neurochem 2013; 128:790-7. [DOI: 10.1111/jnc.12518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 09/18/2013] [Accepted: 10/30/2013] [Indexed: 02/02/2023]
Affiliation(s)
- Takao Hikita
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Akihisa Ohno
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medical Sciences; Nagoya Japan
| | - Haruko Ota
- 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
|
42
|
Van Schepdael A, Ashbourn J, Beard R, Miller J, Geris L. Mechanisms of cell migration in the adult brain: modelling subventricular neurogenesis. Comput Methods Biomech Biomed Engin 2013; 16:1096-105. [DOI: 10.1080/10255842.2013.773979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
Díaz D, Gómez C, Muñoz-Castañeda R, Baltanás F, Alonso JR, Weruaga E. The Olfactory System as a Puzzle: Playing With Its Pieces. Anat Rec (Hoboken) 2013; 296:1383-400. [DOI: 10.1002/ar.22748] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- D. Díaz
- Laboratory of Neuronal Plasticity and Neurorepair; Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca; Salamanca Spain
- Area of Gene and Cell Therapy; Institute of Biomedical Research of Salamanca, IBSAL; Salamanca Spain
| | - C. Gómez
- Laboratory of Neuronal Plasticity and Neurorepair; Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca; Salamanca Spain
- Institute for Molecular and Cell Biology of the Cancer, IBMCC, CSIC-Universidad de Salamanca; Salamanca Spain
| | - R. Muñoz-Castañeda
- Laboratory of Neuronal Plasticity and Neurorepair; Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca; Salamanca Spain
- Area of Gene and Cell Therapy; Institute of Biomedical Research of Salamanca, IBSAL; Salamanca Spain
| | - F. Baltanás
- Laboratory of Neuronal Plasticity and Neurorepair; Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca; Salamanca Spain
- Institute for Molecular and Cell Biology of the Cancer, IBMCC, CSIC-Universidad de Salamanca; Salamanca Spain
| | - J. R. Alonso
- Laboratory of Neuronal Plasticity and Neurorepair; Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca; Salamanca Spain
- Area of Gene and Cell Therapy; Institute of Biomedical Research of Salamanca, IBSAL; Salamanca Spain
- Institute for High Research, Universidad de Tarapacá; Arica Chile
| | - E. Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair; Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca; Salamanca Spain
- Area of Gene and Cell Therapy; Institute of Biomedical Research of Salamanca, IBSAL; Salamanca Spain
| |
Collapse
|
44
|
Cayre M, Courtès S, Martineau F, Giordano M, Arnaud K, Zamaron A, Durbec P. Netrin 1 contributes to vascular remodeling in the subventricular zone and promotes progenitor emigration after demyelination. Development 2013; 140:3107-17. [PMID: 23824572 DOI: 10.1242/dev.092999] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neural stem cells are maintained in the adult brain, sustaining structural and functional plasticity and to some extent participating in brain repair. A thorough understanding of the mechanisms and factors involved in endogenous stem/progenitor cell mobilization is a major challenge in the promotion of spontaneous brain repair. The main neural stem cell niche in the adult brain is the subventricular zone (SVZ). Following demyelination insults, SVZ-derived progenitors act in concert with oligodendrocyte precursors to repopulate the lesion and replace lost oligodendrocytes. Here, we showed robust vascular reactivity within the SVZ after focal demyelination of the corpus callosum in adult mice, together with a remarkable physical association between these vessels and neural progenitors exiting from their niche. Endogenous progenitor cell recruitment towards the lesion was significantly reduced by inhibiting post-lesional angiogenesis in the SVZ using anti-VEGF blocking antibody injections, suggesting a facilitating role of blood vessels for progenitor cell migration towards the lesion. We identified netrin 1 (NTN1) as a key factor upregulated within the SVZ after demyelination and involved in local angiogenesis and progenitor cell migration. Blocking NTN1 expression using a neutralizing antibody inhibited both lesion-induced vascular reactivity and progenitor cell recruitment at the lesion site. We propose a model in which SVZ progenitors respond to a demyelination lesion by NTN1 secretion that both directly promotes cell emigration and contributes to local angiogenesis, which in turn indirectly facilitates progenitor cell emigration from the niche.
Collapse
Affiliation(s)
- Myriam Cayre
- Aix-Marseille Université, IBDM-UMR7288, 13288 Marseille, France.
| | | | | | | | | | | | | |
Collapse
|
45
|
Keasey MP, Kang SS, Lovins C, Hagg T. Inhibition of a novel specific neuroglial integrin signaling pathway increases STAT3-mediated CNTF expression. Cell Commun Signal 2013; 11:35. [PMID: 23693126 PMCID: PMC3691611 DOI: 10.1186/1478-811x-11-35] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 05/13/2013] [Indexed: 11/14/2022] Open
Abstract
Background Ciliary neurotrophic factor (CNTF) expression is repressed in astrocytes by neuronal contact in the CNS and is rapidly induced by injury. Here, we defined an inhibitory integrin signaling pathway. Results The integrin substrates laminin, fibronectin and vitronectin, but not collagen, thrombospondin or fibrinogen, reduced CNTF expression in C6 astroglioma cells. Antibodies against αv and β5, but not α6 or β1, integrin induced CNTF. Together, the ligand and antibody specificity suggests that CNTF is repressed by αvβ5 integrin. Antibodies against Thy1, an abundant neuronal surface protein whose function is unclear, induced CNTF in neuron-astrocyte co-cultures indicating that it is a neuroglial CNTF repressor. Inhibition of the integrin signaling molecule Focal Adhesion Kinase (FAK) or the downstream c-Jun N-terminal kinase (JNK), but not extracellular regulated kinase (ERK) or p38 MAPK, greatly induced CNTF mRNA and protein expression within 4 hours. This selective inhibitory pathway phosphorylated STAT3 on its inhibitory ser-727 residue interfering with activity of the pro-transcription Tyr-705 residue. STAT3 can activate CNTF transcription because it bound to its promoter and FAK antagonist-induced CNTF was reduced by blocking STAT3. Microinjection of FAK inhibitor directly into the brain or spinal cord in adult mice rapidly induced CNTF mRNA and protein expression. Importantly, systemic treatment with FAK inhibitors over 3 days induced CNTF in the subventricular zone and increased neurogenesis. Conclusions Neuron-astroglia contact mediated by integrins serves as a sensor to enable rapid neurotrophic responses and provides a new pharmacological avenue to exploit the neuroprotective properties of endogenous CNTF.
Collapse
|
46
|
Maucksch C, McGregor AL, Yang M, Gordon RJ, Yang M, Connor B. IGF-I redirects doublecortin-positive cell migration in the normal adult rat brain. Neuroscience 2013; 241:106-15. [PMID: 23528977 DOI: 10.1016/j.neuroscience.2013.03.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 02/26/2013] [Accepted: 03/09/2013] [Indexed: 01/05/2023]
Abstract
The migration of subventricular zone (SVZ)-derived neural precursor cells through the rostral migratory stream (RMS) to the olfactory bulb is tightly regulated by local micro-environmental cues. Insulin-like Growth Factor-I (IGF-I) can stimulate the migration of several neuronal cell types and acts as a 'departure' factor in the avian SVZ. To establish whether IGF-I can also act as a migratory factor for adult neuronal precursor cells in vivo, in addition to its well established role in precursor cell proliferation and differentiation, we used AAV2-mediated gene transfer to produce ectopic expression of IGF-I in the normal adult rat striatum. We then assessed whether the expression of IGF-I would recruit SVZ-derived neuronal precursor cells from the RMS into the striatum. Ectopic expression of IGF-I in the normal adult rat brain significantly increased the number of doublecortin (Dcx)-positive cells and the extent of their migration into the striatum 4 and 8 weeks after AAV2-IGF-I injection but did not promote neuronal differentiation. In vitro migration assays confirmed that IGF-I is an inducer of migration and directs SVZ-derived adult neuronal precursor cell migration by both chemotaxis and chemokinesis. These results demonstrate that overexpression of IGF-I in the normal adult rat brain can override the normal cues directing precursor cell migration along the RMS and can redirect precursor cell migration into a non-neurogenic region. Enhanced expression of IGF-I following brain injury may therefore act as a diffusible factor mediating precursor cell migration to areas of neuronal cell damage.
Collapse
Affiliation(s)
- C Maucksch
- Department of Pharmacology & Clinical Pharmacology, School of Medical Sciences, Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | | | | | | | | | | |
Collapse
|
47
|
Quadrato G, Di Giovanni S. Waking up the sleepers: shared transcriptional pathways in axonal regeneration and neurogenesis. Cell Mol Life Sci 2013; 70:993-1007. [PMID: 22899311 PMCID: PMC11113138 DOI: 10.1007/s00018-012-1099-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 06/13/2012] [Accepted: 07/17/2012] [Indexed: 12/26/2022]
Abstract
In the last several years, relevant progress has been made in our understanding of the transcriptional machinery regulating CNS repair after acute injury, such as following trauma or stroke. In order to survive and functionally reconnect to the synaptic network, injured neurons activate an intrinsic rescue program aimed to increase their plasticity. Perhaps, in the attempt to switch back to a plastic and growth-competent state, post-mitotic neurons wake up and re-express a set of transcription factors that are also critical for the regulation of their younger brothers, the neural stem cells. Here, we review and discuss the transcriptional pathways regulating both axonal regeneration and neurogenesis highlighting the connection between the two. Clarification of their common molecular substrate may help simultaneous targeting of both neurogenesis and axonal regeneration with the hope to enhance functional recovery following CNS injury.
Collapse
Affiliation(s)
- Giorgia Quadrato
- Laboratory for NeuroRegeneration and Repair, Center for Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Otfried-Mueller Strasse 27, 72076 Tuebingen, Germany
| | - Simone Di Giovanni
- Laboratory for NeuroRegeneration and Repair, Center for Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Otfried-Mueller Strasse 27, 72076 Tuebingen, Germany
| |
Collapse
|
48
|
Spatiotemporal expression of repulsive guidance molecules (RGMs) and their receptor neogenin in the mouse brain. PLoS One 2013; 8:e55828. [PMID: 23457482 PMCID: PMC3573027 DOI: 10.1371/journal.pone.0055828] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 01/02/2013] [Indexed: 02/07/2023] Open
Abstract
Neogenin has been implicated in a variety of developmental processes such as neurogenesis, neuronal differentiation, apoptosis, migration and axon guidance. Binding of repulsive guidance molecules (RGMs) to Neogenin inhibits axon outgrowth of different neuronal populations. This effect requires Neogenin to interact with co-receptors of the uncoordinated locomotion-5 (Unc5) family to activate downstream Rho signaling. Although previous studies have reported RGM, Neogenin, and/or Unc5 expression, a systematic comparison of RGM and Neogenin expression in the developing nervous system is lacking, especially at later developmental stages. Furthermore, information on RGM and Neogenin expression at the protein level is limited. To fill this void and to gain further insight into the role of RGM-Neogenin signaling during mouse neural development, we studied the expression of RGMa, RGMb, Neogenin and Unc5A-D using in situ hybridization, immunohistochemistry and RGMa section binding. Expression patterns in the primary olfactory system, cortex, hippocampus, habenula, and cerebellum were studied in more detail. Characteristic cell layer-specific expression patterns were detected for RGMa, RGMb, Neogenin and Unc5A-D. Furthermore, strong expression of RGMa, RGMb and Neogenin protein was found on several major axon tracts such as the primary olfactory projections, anterior commissure and fasciculus retroflexus. These data not only hint at a role for RGM-Neogenin signaling during the development of different neuronal systems, but also suggest that Neogenin partners with different Unc5 family members in different systems. Overall, the results presented here will serve as a framework for further dissection of the role of RGM-Neogenin signaling during neural development.
Collapse
|
49
|
Ju XD, Guo Y, Wang NN, Huang Y, Lai MM, Zhai YH, Guo YG, Zhang JH, Cao RJ, Yu HL, Cui L, Li YT, Wang XZ, Ding YQ, Zhu XJ. Both Myosin-10 isoforms are required for radial neuronal migration in the developing cerebral cortex. ACTA ACUST UNITED AC 2013; 24:1259-68. [PMID: 23300110 DOI: 10.1093/cercor/bhs407] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
During embryonic development of the mammalian cerebral cortex, postmitotic cortical neurons migrate radially from the ventricular zone to the cortical plate. Proper migration involves the correct orientation of migrating neurons and the transition from a multipolar to a mature bipolar morphology. Herein, we report that the 2 isoforms of Myosin-10 (Myo10) play distinct roles in the regulation of radial migration in the mouse cortex. We show that the full-length Myo10 (fMyo10) isoform is located in deeper layers of the cortex and is involved in establishing proper migration orientation. We also demonstrate that fMyo10-dependent orientation of radial migration is mediated at least in part by the netrin-1 receptor deleted in colorectal cancer. Moreover, we show that the headless Myo10 (hMyo10) isoform is required for the transition from multipolar to bipolar morphologies in the intermediate zone. Our study reveals divergent functions for the 2 Myo10 isoforms in controlling both the direction of migration and neuronal morphogenesis during radial cortical neuronal migration.
Collapse
Affiliation(s)
- Xing-Da Ju
- Key Laboratory of Molecular Epigenetics, Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130021, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cao N, Feng J, Bai J, Sun L, Li S, Ma J, Wang L. Netrin-1 attenuates the progression of renal dysfunction by inhibiting peritubular capillary loss and hypoxia in 5/6 nephrectomized rats. Kidney Blood Press Res 2012; 36:209-19. [PMID: 23147235 DOI: 10.1159/000343410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND/AIMS The aim of this study was to investigate the effect of netrin-1 on peritubular capillary (PTC) loss and hypoxia in 5/6 nephrectomized (Nx) rats. METHODS Male Sprague-Dawley rats were divided into three groups (n = 10 rats/group): sham-operated rats treated with control adenovirus; 5/6 Nx rats treated with control adenovirus; and 5/6 Nx rats treated with recombinant adenovirus mediated netrin-1 gene (Ad-netrin-1) therapy. Rats were killed 12 weeks after surgery. Blood urea nitrogen (BUN), serum creatinine (Scr) and 24-h urinary albumin excretion rates were measured. Pathological changes in renal tissues were analyzed histologically. The concentration of netrin-1, CD34, and hypoxia-inducible factor-1α (HIF-1α) were analyzed by immunohistochemistry, Western blotting and real-time PCR. RESULTS Renal function and histopathological damage were significantly improved in Adnetrin-1 treated 5/6 Nx rats, compared with rats treated with the control adenovirus in the 5/6 Nx group. Furthermore, Ad-netrin-1 treatment induced a significant increase in renal PTC density, accompanied by a significant decrease in HIF-1α expression. CONCLUSION Adenovirus mediated netrin-1 treatment attenuates PTC damage, relieves tissues hypoxia and improves renal function, thus alleviating renal pathological changes and interstitial fibrosis in 5/6 Nx rats.
Collapse
Affiliation(s)
- Ning Cao
- Department of Nephrology, First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | | | |
Collapse
|