1
|
Fogli M, Nato G, Greulich P, Pinto J, Ribodino M, Valsania G, Peretto P, Buffo A, Luzzati F. Dynamic spatiotemporal activation of a pervasive neurogenic competence in striatal astrocytes supports continuous neurogenesis following injury. Stem Cell Reports 2024; 19:1432-1450. [PMID: 39303706 PMCID: PMC11561465 DOI: 10.1016/j.stemcr.2024.08.006] [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: 03/23/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Adult neural stem cells (NSCs) are conventionally regarded as rare cells restricted to two niches: the subventricular zone (SVZ) and the subgranular zone. Parenchymal astrocytes (ASs) can also contribute to neurogenesis after injury; however, the prevalence, distribution, and behavior of these latent NSCs remained elusive. To tackle these issues, we reconstructed the spatiotemporal pattern of striatal (STR) AS neurogenic activation after excitotoxic lesion in mice. Our results indicate that neurogenic potential is widespread among STR ASs but is focally activated at the lesion border, where it associates with different reactive AS subtypes. In this region, similarly to canonical niches, steady-state neurogenesis is ensured by the continuous stochastic activation of local ASs. Activated ASs quickly return to quiescence, while their progeny transiently expand following a stochastic behavior that features an acceleration in differentiation propensity. Notably, STR AS activation rate matches that of SVZ ASs indicating a comparable prevalence of NSC potential.
Collapse
Affiliation(s)
- Marco Fogli
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Giulia Nato
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Philip Greulich
- School of Mathematical Sciences, University of Southampton, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK
| | - Jacopo Pinto
- Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Marta Ribodino
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Gregorio Valsania
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Paolo Peretto
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Annalisa Buffo
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Federico Luzzati
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy.
| |
Collapse
|
2
|
Crisci I, Bonzano S, Nicolas Z, Dallorto E, Peretto P, Krezel W, De Marchis S. Tamoxifen exerts direct and microglia-mediated effects preventing neuroinflammatory changes in the adult mouse hippocampal neurogenic niche. Glia 2024; 72:1273-1289. [PMID: 38515286 DOI: 10.1002/glia.24526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/23/2024]
Abstract
Tamoxifen-inducible systems are widely used in research to control Cre-mediated gene deletion in genetically modified animals. Beyond Cre activation, tamoxifen also exerts off-target effects, whose consequences are still poorly addressed. Here, we investigated the impact of tamoxifen on lipopolysaccharide (LPS)-induced neuroinflammatory responses, focusing on the neurogenic activity in the adult mouse dentate gyrus. We demonstrated that a four-day LPS treatment led to an increase in microglia, astrocytes and radial glial cells with concomitant reduction of newborn neurons. These effects were counteracted by a two-day tamoxifen pre-treatment. Through selective microglia depletion, we elucidated that both LPS and tamoxifen influenced astrogliogenesis via microglia mediated mechanisms, while the effects on neurogenesis persisted even in a microglia-depleted environment. Notably, changes in radial glial cells resulted from a combination of microglia-dependent and -independent mechanisms. Overall, our data reveal that tamoxifen treatment per se does not alter the balance between adult neurogenesis and astrogliogenesis but does modulate cellular responses to inflammatory stimuli exerting a protective role within the adult hippocampal neurogenic niche.
Collapse
Affiliation(s)
- Isabella Crisci
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
- NICO-Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
- NICO-Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Zinter Nicolas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | - Eleonora Dallorto
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
- NICO-Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
- NICO-Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Wojciech Krezel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | - Silvia De Marchis
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
- NICO-Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| |
Collapse
|
3
|
Zheng XQ, Lin JL, Huang J, Wu T, Song CL. Targeting aging with the healthy skeletal system: The endocrine role of bone. Rev Endocr Metab Disord 2023; 24:695-711. [PMID: 37402956 DOI: 10.1007/s11154-023-09812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 07/06/2023]
Abstract
Aging is an inevitable biological process, and longevity may be related to bone health. Maintaining strong bone health can extend one's lifespan, but the exact mechanism is unclear. Bone and extraosseous organs, including the heart and brain, have complex and precise communication mechanisms. In addition to its load bearing capacity, the skeletal system secretes cytokines, which play a role in bone regulation of extraosseous organs. FGF23, OCN, and LCN2 are three representative bone-derived cytokines involved in energy metabolism, endocrine homeostasis and systemic chronic inflammation levels. Today, advanced research methods provide new understandings of bone as a crucial endocrine organ. For example, gene editing technology enables bone-specific conditional gene knockout models, which allows the study of bone-derived cytokines to be more precise. We systematically evaluated the various effects of bone-derived cytokines on extraosseous organs and their possible antiaging mechanism. Targeting aging with the current knowledge of the healthy skeletal system is a potential therapeutic strategy. Therefore, we present a comprehensive review that summarizes the current knowledge and provides insights for futures studies.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Tong Wu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China.
| |
Collapse
|
4
|
Yeo RW, Zhou OY, Zhong BL, Sun ED, Navarro Negredo P, Nair S, Sharmin M, Ruetz TJ, Wilson M, Kundaje A, Dunn AR, Brunet A. Chromatin accessibility dynamics of neurogenic niche cells reveal defects in neural stem cell adhesion and migration during aging. NATURE AGING 2023; 3:866-893. [PMID: 37443352 PMCID: PMC10353944 DOI: 10.1038/s43587-023-00449-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/02/2023] [Indexed: 07/15/2023]
Abstract
The regenerative potential of brain stem cell niches deteriorates during aging. Yet the mechanisms underlying this decline are largely unknown. Here we characterize genome-wide chromatin accessibility of neurogenic niche cells in vivo during aging. Interestingly, chromatin accessibility at adhesion and migration genes decreases with age in quiescent neural stem cells (NSCs) but increases with age in activated (proliferative) NSCs. Quiescent and activated NSCs exhibit opposing adhesion behaviors during aging: quiescent NSCs become less adhesive, whereas activated NSCs become more adhesive. Old activated NSCs also show decreased migration in vitro and diminished mobilization out of the niche for neurogenesis in vivo. Using tension sensors, we find that aging increases force-producing adhesions in activated NSCs. Inhibiting the cytoskeletal-regulating kinase ROCK reduces these adhesions, restores migration in old activated NSCs in vitro, and boosts neurogenesis in vivo. These results have implications for restoring the migratory potential of NSCs and for improving neurogenesis in the aged brain.
Collapse
Affiliation(s)
- Robin W Yeo
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Olivia Y Zhou
- Department of Genetics, Stanford University, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University, Stanford, CA, USA
| | - Brian L Zhong
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Eric D Sun
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biomedical Informatics Graduate Program, Stanford University, Stanford, CA, USA
| | | | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Mahfuza Sharmin
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Tyson J Ruetz
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Mikaela Wilson
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Intranasal delivery of full-length anti-Nogo-A antibody: A potential alternative route for therapeutic antibodies to central nervous system targets. Proc Natl Acad Sci U S A 2023; 120:e2200057120. [PMID: 36649432 PMCID: PMC9942809 DOI: 10.1073/pnas.2200057120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Antibody delivery to the CNS remains a huge hurdle for the clinical application of antibodies targeting a CNS antigen. The blood-brain barrier and blood-CSF barrier restrict access of therapeutic antibodies to their CNS targets in a major way. The very high amounts of therapeutic antibodies that are administered systemically in recent clinical trials to reach CNS targets are barely viable cost-wise for broad, routine applications. Though global CNS delivery of antibodies can be achieved by intrathecal application, these procedures are invasive. A non-invasive method to bring antibodies into the CNS reliably and reproducibly remains an important unmet need in neurology. In the present study, we show that intranasal application of a mouse monoclonal antibody against the neurite growth-inhibiting and plasticity-restricting membrane protein Nogo-A leads to a rapid transfer of significant amounts of antibody to the brain and spinal cord in intact adult rats. Daily intranasal application for 2 wk of anti-Nogo-A antibody enhanced growth and compensatory sprouting of corticofugal projections and functional recovery in rats after large unilateral cortical strokes. These findings are a starting point for clinical translation for a less invasive route of application of therapeutic antibodies to CNS targets for many neurological indications.
Collapse
|
6
|
The Genomic Architecture of Pregnancy-Associated Plasticity in the Maternal Mouse Hippocampus. eNeuro 2022; 9:ENEURO.0117-22.2022. [PMID: 36239981 PMCID: PMC9522463 DOI: 10.1523/eneuro.0117-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/10/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Pregnancy is associated with extraordinary plasticity in the maternal brain. Studies in humans and other mammals suggest extensive structural and functional remodeling of the female brain during and after pregnancy. However, we understand remarkably little about the molecular underpinnings of this natural phenomenon. To gain insight into pregnancy-associated hippocampal plasticity, we performed single nucleus RNA sequencing (snRNA-seq) and snATAC-seq from the mouse hippocampus before, during, and after pregnancy. We identified cell type-specific transcriptional and epigenetic signatures associated with pregnancy and postpartum adaptation. In addition, we analyzed receptor-ligand interactions and transcription factor (TF) motifs that inform hippocampal cell type identity and provide evidence of pregnancy-associated adaption. In total, these data provide a unique resource of coupled transcriptional and epigenetic data across a dynamic time period in the mouse hippocampus and suggest opportunities for functional interrogation of hormone-mediated plasticity.
Collapse
|
7
|
Wang L, Liu J, Xu J, Zhang W, Wang R. Coupling of GPR30 mediated neurogenesis and protection with astroglial Aromatase-STAT3 signaling in rat hippocampus after global cerebral ischemia. Mol Cell Endocrinol 2021; 535:111394. [PMID: 34274445 DOI: 10.1016/j.mce.2021.111394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/09/2021] [Accepted: 07/11/2021] [Indexed: 11/28/2022]
Abstract
Our previous study revealed that G-protein-coupled estrogen receptor-30 (GPR30) agonist G1 serves as a viable alternative neuroprotectant of 17β-estradiol (E2) to attenuate neuroinflammation and improve cognitive function after global cerebral ischemia (GCI). Aromatase, the key enzyme of E2 biosynthesis, is widely expressed in animal and human brain, and its expression and activity are mediated by selective estrogen receptor modulators. In the present study, we explored the long-term protective and reparative effects of G1 in ovariectomized rats after GCI. We used the aromatase inhibitor letrozole to elucidate whether G1 and brain-derived E2 together induce beneficial effects. Our results showed that G1 administration for 28 days a) significantly increased neurogenesis in the hippocampal sub-granular zone and CA1 regions; b) declined CA1 neuronal impairment in a long-term fashion; c) enhanced expression of synaptic proteins and cognitive function; d) and prevented reactive astrocytes loss, wherein aromatase and brain-derived estrogen levels were markedly increased. Additionally, expression and activation of transducer and activator of transcription 3 (STAT3) were increased in CA1 astrocytes of G1-treated animals. Letrozole abolished all of the observed benefits of G1. Our results suggest that GPR30 activation mediates long-term neuroprotection and neurogenesis in the hippocampus following GCI, with potential mechanism coupling with the activation of astroglial aromatase-STAT3 signaling.
Collapse
Affiliation(s)
- Lu Wang
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China
| | - Jiahao Liu
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China
| | - Jing Xu
- Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China; School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Wenli Zhang
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China
| | - Ruimin Wang
- School of Public Health of North China University of Science and Technology, Tangshan, Hebei, 063210, China; Dementia and Dyscognitive Key Lab, Tangshan, Hebei, 063000, China; International Science & Technology Cooperation Base of Geriatric Medicine, Tangshan, Hebei, 063000, China; School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
8
|
Stehle JH, Sheng Z, Hausmann L, Bechstein P, Weinmann O, Hernesniemi J, Neimat JS, Schwab ME, Zemmar A. Exercise-induced Nogo-A influences rodent motor learning in a time-dependent manner. PLoS One 2021; 16:e0250743. [PMID: 33951058 PMCID: PMC8099082 DOI: 10.1371/journal.pone.0250743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 11/22/2022] Open
Abstract
The adult, mature central nervous system (CNS) has limited plasticity. Physical exercising can counteract this limitation by inducing plasticity and fostering processes such as learning, memory consolidation and formation. Little is known about the molecular factors that govern these mechanisms, and how they are connected with exercise. In this study, we used immunohistochemical and behavioral analyses to investigate how running wheel exercise affects expression of the neuronal plasticity-inhibiting protein Nogo-A in the rat cortex, and how it influences motor learning in vivo. Following one week of exercise, rats exhibited a decrease in Nogo-A levels, selectively in motor cortex layer 2/3, but not in layer 5. Nogo-A protein levels returned to baseline after two weeks of running wheel exercise. In a skilled motor task (forelimb-reaching), administration of Nogo-A function-blocking antibodies over the course of the first training week led to improved motor learning. By contrast, Nogo-A antibody application over two weeks of training resulted in impaired learning. Our findings imply a bimodal, time-dependent function of Nogo-A in exercise-induced neuronal plasticity: While an activity-induced suppression of the plasticity-inhibiting protein Nogo-A appears initially beneficial for enhanced motor learning, presumably by allowing greater plasticity in establishing novel synaptic connections, this process is not sustained throughout continued exercise. Instead, upregulation of Nogo-A over the course of the second week of running wheel exercise in rats implies that Nogo-A is required for consolidation of acquired motor skills during the delayed memory consolidation process, possibly by inhibiting ongoing neuronal morphological reorganization to stabilize established synaptic pathways. Our findings suggest that Nogo-A downregulation allows leaning to occur, i.e. opens a 'learning window', while its later upregulation stabilizes the learnt engrams. These findings underline the importance of appropriately timing of application of Nogo-A antibodies in future clinical trials that aim to foster memory performance while avoiding adverse effects.
Collapse
Affiliation(s)
- Jörg H. Stehle
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
- Dr. Senckenbergische Anatomie, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Zhiyuan Sheng
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Laura Hausmann
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Philipp Bechstein
- Dr. Senckenbergische Anatomie, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Oliver Weinmann
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Juha Hernesniemi
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Joseph S. Neimat
- Department of Neurosurgery, University of Louisville, School of Medicine, Louisville, Kentucky, United States of America
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ajmal Zemmar
- Department of Neurosurgery, Henan Provincial People´s Hospital, Henan University People’s Hospital, Henan University School of Medicine, People’s Hospital of Zhengzhou University, Zhengzhou, China
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
The Implication of Reticulons (RTNs) in Neurodegenerative Diseases: From Molecular Mechanisms to Potential Diagnostic and Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094630. [PMID: 33924890 PMCID: PMC8125174 DOI: 10.3390/ijms22094630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Reticulons (RTNs) are crucial regulatory factors in the central nervous system (CNS) as well as immune system and play pleiotropic functions. In CNS, RTNs are transmembrane proteins mediating neuroanatomical plasticity and functional recovery after central nervous system injury or diseases. Moreover, RTNs, particularly RTN4 and RTN3, are involved in neurodegeneration and neuroinflammation processes. The crucial role of RTNs in the development of several neurodegenerative diseases, including Alzheimer's disease (AD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), or other neurological conditions such as brain injury or spinal cord injury, has attracted scientific interest. Reticulons, particularly RTN-4A (Nogo-A), could provide both an understanding of early pathogenesis of neurodegenerative disorders and be potential therapeutic targets which may offer effective treatment or inhibit disease progression. This review focuses on the molecular mechanisms and functions of RTNs and their potential usefulness in clinical practice as a diagnostic tool or therapeutic strategy.
Collapse
|
10
|
Nakajima C, Sawada M, Sawamoto K. Postnatal neuronal migration in health and disease. Curr Opin Neurobiol 2020; 66:1-9. [PMID: 32717548 DOI: 10.1016/j.conb.2020.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/02/2020] [Indexed: 10/23/2022]
Abstract
Postnatal neuronal migration modulates neuronal circuit formation and function throughout life and is conserved among species. Pathological conditions activate the generation of neuroblasts in the ventricular-subventricular zone (V-SVZ) and promote their migration towards a lesion. However, the neuroblasts generally terminate their migration before reaching the lesion site unless their intrinsic capacity is modified or the environment is improved. It is important to understand which factors impede neuronal migration for functional recovery of the brain. We highlight similarities and differences in the mechanisms of neuroblast migration under physiological and pathological conditions to provide novel insights into endogenous neuronal regeneration.
Collapse
Affiliation(s)
- Chikako Nakajima
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Division of Neural Development and Regeneration, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Division of Neural Development and Regeneration, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.
| |
Collapse
|
11
|
Motahari Z, Moody SA, Maynard TM, LaMantia AS. In the line-up: deleted genes associated with DiGeorge/22q11.2 deletion syndrome: are they all suspects? J Neurodev Disord 2019; 11:7. [PMID: 31174463 PMCID: PMC6554986 DOI: 10.1186/s11689-019-9267-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 04/21/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND 22q11.2 deletion syndrome (22q11DS), a copy number variation (CNV) disorder, occurs in approximately 1:4000 live births due to a heterozygous microdeletion at position 11.2 (proximal) on the q arm of human chromosome 22 (hChr22) (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011). This disorder was known as DiGeorge syndrome, Velo-cardio-facial syndrome (VCFS) or conotruncal anomaly face syndrome (CTAF) based upon diagnostic cardiovascular, pharyngeal, and craniofacial anomalies (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011; Burn et al., J Med Genet 30:822-4, 1993) before this phenotypic spectrum was associated with 22q11.2 CNVs. Subsequently, 22q11.2 deletion emerged as a major genomic lesion associated with vulnerability for several clinically defined behavioral deficits common to a number of neurodevelopmental disorders (Fernandez et al., Principles of Developmental Genetics, 2015; Robin and Shprintzen, J Pediatr 147:90-6, 2005; Schneider et al., Am J Psychiatry 171:627-39, 2014). RESULTS The mechanistic relationships between heterozygously deleted 22q11.2 genes and 22q11DS phenotypes are still unknown. We assembled a comprehensive "line-up" of the 36 protein coding loci in the 1.5 Mb minimal critical deleted region on hChr22q11.2, plus 20 protein coding loci in the distal 1.5 Mb that defines the 3 Mb typical 22q11DS deletion. We categorized candidates based upon apparent primary cell biological functions. We analyzed 41 of these genes that encode known proteins to determine whether haploinsufficiency of any single 22q11.2 gene-a one gene to one phenotype correspondence due to heterozygous deletion restricted to that locus-versus complex multigenic interactions can account for single or multiple 22q11DS phenotypes. CONCLUSIONS Our 22q11.2 functional genomic assessment does not support current theories of single gene haploinsufficiency for one or all 22q11DS phenotypes. Shared molecular functions, convergence on fundamental cell biological processes, and related consequences of individual 22q11.2 genes point to a matrix of multigenic interactions due to diminished 22q11.2 gene dosage. These interactions target fundamental cellular mechanisms essential for development, maturation, or homeostasis at subsets of 22q11DS phenotypic sites.
Collapse
Affiliation(s)
- Zahra Motahari
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Sally Ann Moody
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Thomas Michael Maynard
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Anthony-Samuel LaMantia
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| |
Collapse
|
12
|
Cerrato V, Parmigiani E, Figueres-Oñate M, Betizeau M, Aprato J, Nanavaty I, Berchialla P, Luzzati F, de’Sperati C, López-Mascaraque L, Buffo A. Multiple origins and modularity in the spatiotemporal emergence of cerebellar astrocyte heterogeneity. PLoS Biol 2018; 16:e2005513. [PMID: 30260948 PMCID: PMC6178385 DOI: 10.1371/journal.pbio.2005513] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 10/09/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022] Open
Abstract
The morphological, molecular, and functional heterogeneity of astrocytes is under intense scrutiny, but how this diversity is ontogenetically achieved remains largely unknown. Here, by quantitative in vivo clonal analyses and proliferation studies, we demonstrate that the major cerebellar astrocyte types emerge according to an unprecedented and remarkably orderly developmental program comprising (i) a time-dependent decline in both clone size and progenitor multipotency, associated with clone allocation first to the hemispheres and then to the vermis(ii) distinctive clonal relationships among astrocyte types, revealing diverse lineage potentials of embryonic and postnatal progenitors; and (iii) stereotyped clone architectures and recurrent modularities that correlate to layer-specific dynamics of postnatal proliferation/differentiation. In silico simulations indicate that the sole presence of a unique multipotent progenitor at the source of the whole astrogliogenic program is unlikely and rather suggest the involvement of additional committed components. Astrocytes are abundant cells of the brain essential to support and shape neuronal activity. They can be grouped in different subclasses based on their remarkable variety of morphologies, molecular profiles, and specialized functions. Although different astrocyte types likely display specialized interactions with distinct neuron categories, the different classes of astrocytes have only partially been unmasked. How astrocyte heterogeneity is ontogenetically achieved remains largely unknown. Here we approached this question by studying the development of the main astrocyte types of the cerebellum. The reconstruction of developmental lineages in the mouse embryo combined with proliferation studies and computational modeling demonstrate that cerebellar astrocyte types emerge according to an unprecedented and remarkably orderly developmental program. Embryonic progenitor cells produce either only a single astrocyte type or more types. These distinct astrocyte lineages display stereotyped architectures and recurrent modularities. Moreover, the generation of astrocytes follows a well-defined spatiotemporal pattern, defined by a time-dependent allocation of astrocytes to distinct cerebellar territories and an inside-out sequence of differentiation, coupled with a decline over time in both progenitor amplification and capability to produce distinct astrocyte types. These results provide the first evidence that an ontogenetic program, tightly regulated in space and time, determines astrocyte heterogeneity.
Collapse
Affiliation(s)
- Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Turin, Italy
| | - Elena Parmigiani
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
| | - María Figueres-Oñate
- Department of Molecular, Cellular, and Developmental Neurobiology, Cajal Institute -CSIC-, Spanish National Research Council, Madrid, Spain
| | - Marion Betizeau
- Brain Research Institute, University of Zurich Irchel, Zurich, Switzerland
| | - Jessica Aprato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
| | - Ishira Nanavaty
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
| | - Paola Berchialla
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Federico Luzzati
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Turin, Italy
- Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Claudio de’Sperati
- Laboratory of Action, Perception and Cognition, Vita-Salute San Raffaele University, Milan, Italy
- Experimental Psychology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura López-Mascaraque
- Department of Molecular, Cellular, and Developmental Neurobiology, Cajal Institute -CSIC-, Spanish National Research Council, Madrid, Spain
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Turin, Italy
- * E-mail:
| |
Collapse
|
13
|
Engler A, Rolando C, Giachino C, Saotome I, Erni A, Brien C, Zhang R, Zimber-Strobl U, Radtke F, Artavanis-Tsakonas S, Louvi A, Taylor V. Notch2 Signaling Maintains NSC Quiescence in the Murine Ventricular-Subventricular Zone. Cell Rep 2018; 22:992-1002. [PMID: 29386140 DOI: 10.1016/j.celrep.2017.12.094] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 06/12/2017] [Accepted: 12/22/2017] [Indexed: 12/21/2022] Open
Abstract
Neurogenesis continues in the ventricular-subventricular zone (V-SVZ) of the adult forebrain from quiescent neural stem cells (NSCs). V-SVZ NSCs are a reservoir for new olfactory bulb (OB) neurons that migrate through the rostral migratory stream (RMS). To generate neurons, V-SVZ NSCs need to activate and enter the cell cycle. The mechanisms underlying NSC transition from quiescence to activity are poorly understood. We show that Notch2, but not Notch1, signaling conveys quiescence to V-SVZ NSCs by repressing cell-cycle-related genes and neurogenesis. Loss of Notch2 activates quiescent NSCs, which proliferate and generate new neurons of the OB lineage. Notch2 deficiency results in accelerated V-SVZ NSC exhaustion and an aging-like phenotype. Simultaneous loss of Notch1 and Notch2 resembled the total loss of Rbpj-mediated canonical Notch signaling; thus, Notch2 functions are not compensated in NSCs, and Notch2 is indispensable for the maintenance of NSC quiescence in the adult V-SVZ.
Collapse
Affiliation(s)
- Anna Engler
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Chiara Rolando
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Claudio Giachino
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Ichiko Saotome
- Departments of Neurosurgery and Neuroscience, Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Andrea Erni
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Callum Brien
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland; School of Science and Technology, Nottingham Trent University, Clifton Campus, NG11 8NS Nottingham, UK
| | - Runrui Zhang
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Ursula Zimber-Strobl
- Department of Gene Vectors, Helmholtz Zentrum München, Marchioninistrasse 25, 81377 Munich, Germany
| | - Freddy Radtke
- EPFL SV ISREC UPRAD, SV 2534 (Bâtiment SV), Station 19, 1015 Lausanne, Switzerland
| | | | - Angeliki Louvi
- Departments of Neurosurgery and Neuroscience, Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland.
| |
Collapse
|
14
|
Song J, Kim OY. Perspectives in Lipocalin-2: Emerging Biomarker for Medical Diagnosis and Prognosis for Alzheimer's Disease. Clin Nutr Res 2018; 7:1-10. [PMID: 29423384 PMCID: PMC5796918 DOI: 10.7762/cnr.2018.7.1.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/16/2017] [Accepted: 01/08/2018] [Indexed: 01/23/2023] Open
Abstract
Lipocalin-2 (LCN2), a secreted glycoprotein belonging to the lipocalin superfamily was reported to participate in various biological processes including cell migration, cell survival, inflammatory responses, and insulin sensitivity. LCN2 is expressed in the multiple tissues such as kidney, liver, uterus, and bone marrow. The receptors for LCN2 were additionally found in microglia, astrocytes, epithelial cells, and neurons, but the role of LCN2 in the central nervous system (CNS) has not been fully understood yet. Recently, in vitro, in vivo, and clinical studies reported the association between LCN2 and the risk of Alzheimer's disease (AD). Here, we reviewed the significant evidences showing that LCN2 contributes to the onset and progression of AD. It may suggest that the manipulation of LCN2 in the CNS would be a crucial target for regulation of the pathogenesis and risk of AD.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, Korea.,Human Life Research Center, Dong-A University, Busan 49315, Korea
| | - Oh Yoen Kim
- Human Life Research Center, Dong-A University, Busan 49315, Korea.,Department of Food Science and Nutrition, Brain Busan 21 Project, Dong-A University, Busan 49315, Korea
| |
Collapse
|
15
|
Lu Y, Hsiang F, Chang JH, Yao XQ, Zhao H, Zou HY, Wang L, Zhang QX. Houshiheisan and its components promote axon regeneration after ischemic brain injury. Neural Regen Res 2018; 13:1195-1203. [PMID: 30028327 PMCID: PMC6065233 DOI: 10.4103/1673-5374.235031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Houshiheisan, a classic prescription in traditional Chinese medicine, contains Flos Chrysanthemi, Radix Saposhnikoviae, Ramulus Cinnamomi, Rhizoma Chuanxiong, Radix et Rhizoma Asari, Radix Platycodonis, Rhizoma Atractylodis macrocephalae, Poria, Rhizoma Zingiberis, Radix Angelicae sinensis, Radix et Rhizoma Ginseng, Radix Scutellariae and Concha Ostreae. According to traditional Chinese medicine theory, Flos Chrysanthemi, Radix Saposhnikoviae, Ramulus Cinnamomi, Rhizoma Chuanxiong, Radix et Rhizoma Asari and Radix Platycodonis are wind-dispelling drugs; Rhizoma Atractylodis macrocephalae, Poria, Rhizoma Zingiberis, Radix Angelicae sinensis and Radix et Rhizoma Ginseng are deficiency-nourishing drugs. A large number of randomized controlled trials have shown that Houshiheisan is effective in treating stroke, but its mechanism of action is unknown. Axonal remodeling is an important mechanism in neural protection and regeneration. Therefore, this study explored the effect and mechanism of action of Houshiheisan on the repair of axons after cerebral ischemia. Rat models of focal cerebral ischemia were established by ligating the right middle cerebral artery. At 6 hours after model establishment, rats were intragastrically administered 10.5 g/kg Houshiheisan or 7.7 g/kg wind-dispelling drug or 2.59 g/kg deficiency-nourishing drug. These medicines were intragastrically administered as above every 24 hours for 7 consecutive days. Houshiheisan, and its wind-dispelling and deficiency-nourishing components reduced the neurological deficit score and ameliorated axon and neuron lesions after cerebral ischemia. Furthermore, Houshiheisan, and its wind-dispelling and deficiency-nourishing components decreased the expression of proteins that inhibit axonal remodeling: amyloid precursor protein, neurite outgrowth inhibitor protein A (Nogo-A), Rho family small GTPase A (RhoA) and Rho-associated kinase 2 (Rock2), and increased the expression of growth associated protein-43, microtubule-associated protein-2, netrin-1, Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division cycle 42 (Cdc42). The effect of Houshiheisan was stronger than wind-dispelling drugs or deficiency-nourishing drugs alone. In conclusion, Houshiheisan, and wind-dispelling and deficiency-nourishing drugs promote the repair of axons and nerve regeneration after cerebral ischemia through Nogo-A/RhoA/Rock2 and Netrin-1/Rac1/Cdc42 signaling pathways. These effects are strongest with Houshiheisan.
Collapse
Affiliation(s)
- Yue Lu
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Flora Hsiang
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Jia-Hui Chang
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Xiao-Quan Yao
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Hai-Yan Zou
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Qiu-Xia Zhang
- School of Traditional Chinese Medicine, Capital Medical University; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| |
Collapse
|
16
|
Kempf A, Boda E, Kwok JC, Fritz R, Grande V, Kaelin AM, Ristic Z, Schmandke A, Schmandke A, Tews B, Fawcett JW, Pertz O, Buffo A, Schwab ME. Control of Cell Shape, Neurite Outgrowth, and Migration by a Nogo-A/HSPG Interaction. Dev Cell 2017; 43:24-34.e5. [DOI: 10.1016/j.devcel.2017.08.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 07/04/2017] [Accepted: 08/21/2017] [Indexed: 11/16/2022]
|
17
|
Shepherd DJ, Tsai SY, Cappucci SP, Wu JY, Farrer RG, Kartje GL. The Subventricular Zone Response to Stroke Is Not a Therapeutic Target of Anti-Nogo-A Immunotherapy. J Neuropathol Exp Neurol 2017; 76:683-696. [DOI: 10.1093/jnen/nlx050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Daniel J. Shepherd
- From the Loyola University Health Sciences Division, Maywood, Illinois (DJS, SPC, GLK); and Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, Illinois (DJS, S-YT, SPC, JYW, RGF, GLK)
| | - Shih-Yen Tsai
- From the Loyola University Health Sciences Division, Maywood, Illinois (DJS, SPC, GLK); and Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, Illinois (DJS, S-YT, SPC, JYW, RGF, GLK)
| | - Stefanie P. Cappucci
- From the Loyola University Health Sciences Division, Maywood, Illinois (DJS, SPC, GLK); and Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, Illinois (DJS, S-YT, SPC, JYW, RGF, GLK)
| | - Joanna Y. Wu
- From the Loyola University Health Sciences Division, Maywood, Illinois (DJS, SPC, GLK); and Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, Illinois (DJS, S-YT, SPC, JYW, RGF, GLK)
| | - Robert G. Farrer
- From the Loyola University Health Sciences Division, Maywood, Illinois (DJS, SPC, GLK); and Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, Illinois (DJS, S-YT, SPC, JYW, RGF, GLK)
| | - Gwendolyn L. Kartje
- From the Loyola University Health Sciences Division, Maywood, Illinois (DJS, SPC, GLK); and Edward Hines Jr. Veterans Affairs Hospital, Research Service, Hines, Illinois (DJS, S-YT, SPC, JYW, RGF, GLK)
| |
Collapse
|
18
|
Hou XQ, Wang L, Wang FG, Zhao XM, Zhang HT. Combination of RNA Interference and Stem Cells for Treatment of Central Nervous System Diseases. Genes (Basel) 2017; 8:genes8050135. [PMID: 28481269 PMCID: PMC5448009 DOI: 10.3390/genes8050135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/31/2022] Open
Abstract
RNA interference (RNAi), including microRNAs, is an important player in the mediation of differentiation and migration of stem cells via target genes. It is used as a potential strategy for gene therapy for central nervous system (CNS) diseases. Stem cells are considered vectors of RNAi due to their capacity to deliver RNAi to other cells. In this review, we discuss the recent advances in studies of RNAi pathways in controlling neuronal differentiation and migration of stem cells. We also highlight the utilization of a combination of RNAi and stem cells in treatment of CNS diseases.
Collapse
Affiliation(s)
- Xue-Qin Hou
- Institute of Pharmacology, Taishan Medical University, Taian 271016, Shandong, China.
| | - Lei Wang
- Institute of Pharmacology, Taishan Medical University, Taian 271016, Shandong, China.
| | - Fu-Gang Wang
- Institute of Pharmacology, Taishan Medical University, Taian 271016, Shandong, China.
| | - Xiao-Min Zhao
- Institute of Pharmacology, Taishan Medical University, Taian 271016, Shandong, China.
| | - Han-Ting Zhang
- Institute of Pharmacology, Taishan Medical University, Taian 271016, Shandong, China.
- Departments of Behavioral Medicine & Psychiatry and Physiology & Pharmacology, Blanchette Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| |
Collapse
|
19
|
McAdams RM, McPherson RJ, Kapur RP, Juul SE. Focal Brain Injury Associated with a Model of Severe Hypoxic-Ischemic Encephalopathy in Nonhuman Primates. Dev Neurosci 2017; 39:107-123. [PMID: 28343228 DOI: 10.1159/000456658] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 01/17/2017] [Indexed: 01/08/2023] Open
Abstract
Worldwide, hypoxic-ischemic encephalopathy (HIE) is a major cause of neonatal mortality and morbidity. To better understand the mechanisms contributing to brain injury and improve outcomes in neonates with HIE, better preclinical animal models that mimic the clinical situation following birth asphyxia in term newborns are needed. In an effort to achieve this goal, we modified our nonhuman primate model of HIE induced by in utero umbilical cord occlusion (UCO) to include postnatal hypoxic episodes, in order to simulate apneic events in human neonates with HIE. We describe a cohort of 4 near-term fetal Macaca nemestrina that underwent 18 min of in utero UCO, followed by cesarean section delivery, resuscitation, and subsequent postnatal mechanical ventilation, with exposure to intermittent daily hypoxia (3 min, 8% O2 3-8 times daily for 3 days). After delivery, all animals demonstrated severe metabolic acidosis (pH 7 ± 0.12; mean ± SD) and low APGAR scores (<5 at 10 min of age). Three of 4 animals had both electrographic and clinical seizures. Serial blood samples were collected and plasma metabolites were determined by 2-dimensional gas chromatography coupled with time-of-flight mass spectrometry (GC × GC-TOFMS). The 4 UCO animals and a single nonasphyxiated animal (delivered by cesarean section but without exposure to UCO or prolonged sedation) underwent brain magnetic resonance imaging (MRI) on day 8 of life. Thalamic injury was present on MRI in 3 UCO animals, but not in the control animal. Following necropsy on day 8, brain histopathology revealed neuronal injury/loss and gliosis in portions of the ventrolateral thalamus in all 4 UCO, with 2 animals also demonstrating putamen/globus pallidus involvement. In addition, all 4 UCO animals demonstrated brain stem gliosis, with neuronal loss present in the midbrain, pons, and lateral medulla in 3 of 4 animals. Transmission electron microscopy imaging of the brain tissues was performed, which demonstrated ultrastructural white matter abnormalities, characterized by perinuclear vacuolation and axonal dilation, in 3 of 4 animals. Immunolabeling of Nogo-A, a negative regulator of neuronal growth, was not increased in the injured brains compared to 2 control animals. Using GC × GC-TOFMS, we identified metabolites previously recognized as potential biomarkers of perinatal asphyxia. The basal ganglia-thalamus-brain stem injury produced by UCO is consistent with the deep nuclear/brainstem injury pattern seen in human neonates after severe, abrupt hypoxic-ischemic insults. The UCO model permits timely detection of biomarkers associated with specific patterns of neonatal brain injury, and it may ultimately be useful for validating therapeutic strategies to treat neonatal HIE.
Collapse
Affiliation(s)
- Ryan M McAdams
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
20
|
Parolisi R, Cozzi B, Bonfanti L. Non-neurogenic SVZ-like niche in dolphins, mammals devoid of olfaction. Brain Struct Funct 2017; 222:2625-2639. [PMID: 28238073 DOI: 10.1007/s00429-016-1361-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/22/2016] [Indexed: 11/29/2022]
Abstract
Adult neurogenesis has been implicated in brain plasticity and brain repair. In mammals, it is mostly restricted to specific brain regions and specific physiological functions. The function and evolutionary history of mammalian adult neurogenesis has been elusive so far. The largest neurogenic site in mammals (subventricular zone, SVZ) generates neurons destined to populate the olfactory bulb. The SVZ neurogenic activity appears to be related to the dependence of the species on olfaction since it occurs at high rates throughout life in animals strongly dependent on this function for their survival. Indeed, it dramatically decreases in humans, who do not depend so much on it. This study investigates whether the SVZ neurogenic site exists in mammals devoid of olfaction and olfactory brain structures, such as dolphins. Our results demonstate that a small SVZ-like region persists in these aquatic mammals. However, this region seems to have lost its neurogenic capabilities since neonatal stages. In addition, instead of the typical newly generated neuroblasts, some mature neurons were observed in the dolphin SVZ. Since cetaceans evolved from terrestrial ancestors, non-neurogenic SVZ may indicate extinction of adult neurogenesis in the absence of olfactory function, with the retention of an SVZ-like anatomical region either vestigial or of still unknown role.
Collapse
Affiliation(s)
- Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy.,Department of Veterinary Sciences, University of Turin, Via Leonardo da Vinci, 44, 10095, Grugliasco, TO, Italy
| | - Bruno Cozzi
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy. .,Department of Veterinary Sciences, University of Turin, Via Leonardo da Vinci, 44, 10095, Grugliasco, TO, Italy.
| |
Collapse
|
21
|
Llorens-Bobadilla E, Martin-Villalba A. Adult NSC diversity and plasticity: the role of the niche. Curr Opin Neurobiol 2016; 42:68-74. [PMID: 27978480 DOI: 10.1016/j.conb.2016.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022]
Abstract
Adult somatic stem cells are generally defined as cells with the ability to differentiate into multiple different lineages and to self-renew during long periods of time. These features were long presumed to be represented in one single tissue-specific stem cell. Recent development of single-cell technologies reveals the existence of diversity in fate and activation state of somatic stem cells within the blood, skin and intestinal compartments [1] but also in the adult brain. Here we review how recent advances have expanded our view of neural stem cells (NSCs) as a diverse pool of cells and how the specialized microenvironment in which they reside acts to maintain this diversity. In addition, we discuss the plasticity of the system in the injured brain.
Collapse
Affiliation(s)
- Enric Llorens-Bobadilla
- Department of Molecular Neurobiology, German Cancer Research Center (DFKZ), Heidelberg, Germany.
| | - Ana Martin-Villalba
- Department of Molecular Neurobiology, German Cancer Research Center (DFKZ), Heidelberg, Germany.
| |
Collapse
|
22
|
Shepherd DJ, Tsai SY, O'Brien TE, Farrer RG, Kartje GL. Anti-Nogo-A Immunotherapy Does Not Alter Hippocampal Neurogenesis after Stroke in Adult Rats. Front Neurosci 2016; 10:467. [PMID: 27803646 PMCID: PMC5067305 DOI: 10.3389/fnins.2016.00467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/28/2016] [Indexed: 12/30/2022] Open
Abstract
Ischemic stroke is a leading cause of adult disability, including cognitive impairment. Our laboratory has previously shown that treatment with function-blocking antibodies against the neurite growth inhibitory protein Nogo-A promotes functional recovery after stroke in adult and aged rats, including enhancing spatial memory performance, for which the hippocampus is critically important. Since spatial memory has been linked to hippocampal neurogenesis, we investigated whether anti-Nogo-A treatment increases hippocampal neurogenesis after stroke. Adult rats were subject to permanent middle cerebral artery occlusion followed 1 week later by 2 weeks of antibody treatment. Cellular proliferation in the dentate gyrus was quantified at the end of treatment, and the number of newborn neurons was determined at 8 weeks post-stroke. Treatment with both anti-Nogo-A and control antibodies stimulated the accumulation of new microglia/macrophages in the dentate granule cell layer, but neither treatment increased cellular proliferation or the number of newborn neurons above stroke-only levels. These results suggest that anti-Nogo-A immunotherapy does not increase post-stroke hippocampal neurogenesis.
Collapse
Affiliation(s)
- Daniel J Shepherd
- Neuroscience Institute, Loyola University Chicago Health Sciences DivisionMaywood, IL, USA; Research Service, Edward Hines Jr. VA HospitalHines, IL, USA
| | - Shih-Yen Tsai
- Research Service, Edward Hines Jr. VA Hospital Hines, IL, USA
| | - Timothy E O'Brien
- Department of Mathematics and Statistics, Loyola University Chicago Chicago, IL, USA
| | - Robert G Farrer
- Research Service, Edward Hines Jr. VA Hospital Hines, IL, USA
| | - Gwendolyn L Kartje
- Neuroscience Institute, Loyola University Chicago Health Sciences DivisionMaywood, IL, USA; Research Service, Edward Hines Jr. VA HospitalHines, IL, USA; Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago Health Sciences DivisionMaywood, IL, USA
| |
Collapse
|
23
|
Fan TK, Gundimeda U, Mack WJ, Gopalakrishna R. Counteraction of Nogo-A and axonal growth inhibitors by green tea polyphenols and other natural products. Neural Regen Res 2016; 11:545-6. [PMID: 27212904 PMCID: PMC4870900 DOI: 10.4103/1673-5374.180729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Tiffany K Fan
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Usha Gundimeda
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - William J Mack
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rayudu Gopalakrishna
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
24
|
Cetin I, Tezdig I, Tarakcioglu MC, Kadak MT, Demirel OF, Ozer OF. Serum levels of glial fibrillary acidic protein and Nogo-A in children with autism spectrum disorders. Biomarkers 2016; 21:614-8. [DOI: 10.3109/1354750x.2016.1171901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ihsan Cetin
- Department of Nutrition and Dietetics, School of Health, Batman University, Batman, Turkey
| | - Ihsan Tezdig
- Department of Chemistry, Institute of Science, Batman University, Batman, Turkey
| | - Mahmut Cem Tarakcioglu
- Clinic of Child and Adolescent Psychiatry, Bakirköy Dr Sadi Konuk Training and Educatin Hospital, Istanbul University, İstanbul, Turkey
| | - Muhammed Tayyib Kadak
- Department of Child and Adolescent Psychiatry, Cerrahpaşa School of Medicine, Istanbul University, İstanbul, Turkey
| | - Omer Faruk Demirel
- Department of Psychiatry, Cerrahpaşa School of Medicine, Istanbul University, İstanbul, Turkey
| | - Omer Faruk Ozer
- Department of Biochemistry, BezmiAlem Vakif University, İstanbul, Turkey
| |
Collapse
|
25
|
ZHONG JIANBIN, LI XIE, WAN LIMEI, CHEN ZHIBANG, ZHONG SIMIN, XIAO SONGHUA, YAN ZHENGWEN. Knockdown of NogoA prevents MPP+-induced neurotoxicity in PC12 cells via the mTOR/STAT3 signaling pathway. Mol Med Rep 2015; 13:1427-33. [DOI: 10.3892/mmr.2015.4637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 11/17/2015] [Indexed: 11/06/2022] Open
|
26
|
BRYUKHOVETSKIY IGOR, BRYUKHOVETSKIY ANDREY, KHOTIMCHENKO YURI, MISCHENKO POLINA. Novel cellular and post-genomic technologies in the treatment of glioblastoma multiforme (Review). Oncol Rep 2015; 35:639-48. [DOI: 10.3892/or.2015.4404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/17/2015] [Indexed: 11/05/2022] Open
|
27
|
Fang Y, Yan J, Li C, Zhou X, Yao L, Pang T, Yan M, Zhang L, Mao L, Liao H. The Nogo/Nogo Receptor (NgR) Signal Is Involved in Neuroinflammation through the Regulation of Microglial Inflammatory Activation. J Biol Chem 2015; 290:28901-14. [PMID: 26472924 DOI: 10.1074/jbc.m115.678326] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Indexed: 01/19/2023] Open
Abstract
Microglia have been proposed to play a pivotal role in the inflammation response of the CNS by expressing a range of proinflammatory enzymes and cytokines under pathological stimulus. Our previous study has confirmed that Nogo receptor (NgR), an axon outgrowth inhibition receptor, is also expressed on microglia and regulates cell adhesion and migration behavior in vitro. In the present study, we further investigated the proinflammatory effects and possible mechanisms of Nogo on microglia in vitro. In this study, Nogo peptide, Nogo-P4, a 25-amino acid core inhibitory peptide sequence of Nogo-66, was used. We found that Nogo-P4 was able to induce the expression of inducible nitric-oxide synthase and cyclooxygenase-2 and the release of proinflammatory cytokines, including IL-1β, TNF-α, NO, and prostaglandin E2 in microglia, which could be reversed by NEP1-40 (Nogo-66(1-40) antagonist peptide), phosphatidylinositol-specificphospholipase C, or NgR siRNA treatment. After Nogo-P4 stimulated microglia, the phosphorylation levels of NF-κB and STAT3 were increased obviously, which further mediated microglia expressing proinflammatory factors induced by Nogo-P4. Taken together, we concluded that Nogo peptide could directly take part in CNS inflammatory process by influencing the expression of proinflammatory factors in microglia, which were related to the NF-κB and STAT3 signal pathways. Besides neurite outgrowth restriction, the Nogo/NgR signal might be involved in multiple processes in various inflammation-associated CNS diseases.
Collapse
Affiliation(s)
- Yinquan Fang
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Jun Yan
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Chenhui Li
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Xiao Zhou
- the Department of Biophysics, Saarland University, Homburg 66421, Germany, and
| | - Lemeng Yao
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Tao Pang
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Ming Yan
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Luyong Zhang
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China
| | - Lei Mao
- the Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, China
| | - Hong Liao
- From the Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, 24 Tongjiaxiang Street, Nanjing 210009, China,
| |
Collapse
|
28
|
Seiler S, Di Santo S, Widmer HR. Non-canonical actions of Nogo-A and its receptors. Biochem Pharmacol 2015; 100:28-39. [PMID: 26348872 DOI: 10.1016/j.bcp.2015.08.113] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/31/2015] [Indexed: 12/13/2022]
Abstract
Nogo-A is a myelin associated protein and one of the most potent neurite growth inhibitors in the central nervous system. Interference with Nogo-A signaling has thus been investigated as therapeutic target to promote functional recovery in CNS injuries. Still, the finding that Nogo-A presents a fairly ubiquitous expression in many types of neurons in different brain regions, in the eye and even in the inner ear suggests for further functions besides the neurite growth repression. Indeed, a growing number of studies identified a variety of functions including regulation of neuronal stem cells, modulation of microglial activity, inhibition of angiogenesis and interference with memory formation. Aim of the present commentary is to draw attention on these less well-known and sometimes controversial roles of Nogo-A. Furthermore, we are addressing the role of Nogo-A in neuropathological conditions such as ischemic stroke, schizophrenia and neurodegenerative diseases.
Collapse
Affiliation(s)
- Stefanie Seiler
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University Hospital Bern and University of Bern, CH-3010 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University Hospital Bern and University of Bern, CH-3010 Bern, Switzerland
| | - Hans Rudolf Widmer
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University Hospital Bern and University of Bern, CH-3010 Bern, Switzerland.
| |
Collapse
|
29
|
Peng WS, Qi C, Zhang H, Gao ML, Wang H, Ren F, Li XQ. Distribution of paired immunoglobulin-like receptor B in the nervous system related to regeneration difficulties after unilateral lumbar spinal cord injury. Neural Regen Res 2015; 10:1139-46. [PMID: 26330840 PMCID: PMC4541248 DOI: 10.4103/1673-5374.160111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2015] [Indexed: 12/21/2022] Open
Abstract
Paired immunoglobulin-like receptor B (PirB) is a functional receptor of myelin-associated inhibitors for axonal regeneration and synaptic plasticity in the central nervous system, and thus suppresses nerve regeneration. The regulatory effect of PirB on injured nerves has received a lot of attention. To better understand nerve regeneration inability after spinal cord injury, this study aimed to investigate the distribution of PirB (via immunofluorescence) in the central nervous system and peripheral nervous system 10 days after injury. Immunoreactivity for PirB increased in the dorsal root ganglia, sciatic nerves, and spinal cord segments. In the dorsal root ganglia and sciatic nerves, PirB was mainly distributed along neuronal and axonal membranes. PirB was found to exhibit a diffuse, intricate distribution in the dorsal and ventral regions. Immunoreactivity for PirB was enhanced in some cortical neurons located in the bilateral precentral gyri. Overall, the findings suggest a pattern of PirB immunoreactivity in the nervous system after unilateral spinal transection injury, and also indicate that PirB may suppress repair after injury.
Collapse
Affiliation(s)
- Wan-Shu Peng
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Chao Qi
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Zhang
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mei-Ling Gao
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hong Wang
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Fei Ren
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xia-Qing Li
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
30
|
Sui YP, Zhang XX, Lu JL, Sui F. New Insights into the Roles of Nogo-A in CNS Biology and Diseases. Neurochem Res 2015; 40:1767-85. [PMID: 26266872 DOI: 10.1007/s11064-015-1671-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/15/2015] [Accepted: 07/17/2015] [Indexed: 12/22/2022]
Abstract
Nogos have become a hot topic for its well-known number Nogo-A's big role in clinical matters. It has been recognized that the expression of Nogo-A and the receptor NgR1 inhibit the neuron's growth after CNS injuries or the onset of the MS. The piling evidence supports the notion that the Nogo-A is also involved in the synaptic plasticity, which was shown to negatively regulate the strength of synaptic transmission. The occurrence of significant schizophrenia-like behavioral phenotypes in Nogo-A KO rats also added strong proof to this conclusion. This review mainly focuses on the structure of Nogo-A and its corresponding receptor-NgR1, its intra- and extra-cellular signaling, together with its major physiological functions such as regulation of migration and distribution and its related diseases like stroke, AD, ALS and so on.
Collapse
Affiliation(s)
- Yun-Peng Sui
- Institute of Chinese Material Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | | | | | | |
Collapse
|
31
|
Diverse functional roles of lipocalin-2 in the central nervous system. Neurosci Biobehav Rev 2015; 49:135-56. [DOI: 10.1016/j.neubiorev.2014.12.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 12/16/2022]
|
32
|
Ramasamy S, Yu F, Hong Yu Y, Srivats H, Dawe GS, Ahmed S. NogoR1 and PirB signaling stimulates neural stem cell survival and proliferation. Stem Cells 2015; 32:1636-48. [PMID: 24449409 DOI: 10.1002/stem.1645] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/11/2013] [Indexed: 11/07/2022]
Abstract
Neural stem cells (NSCs) and neural progenitors (NPs) in the mammalian neocortex give rise to the main cell types of the nervous system. The biological behavior of these NSCs and NPs is regulated by extracellular niche derived autocrine-paracrine signaling factors on a developmental timeline. Our previous reports [Plos One 2010;5:e15341; J Neurochem 2011;117:565-578] have shown that chondroitin sulfate proteoglycan and ApolipoproteinE are autocrine-paracrine survival factors for NSCs. NogoA, a myelin related protein, is expressed in the cortical ventricular zones where NSCs reside. However, the functional role of Nogo signaling proteins in NSC behavior is not completely understood. In this study, we show that NogoA receptors, NogoR1 and PirB, are expressed in the ventricular zone where NSCs reside between E10.5 and 14.5 but not at E15.5. Nogo ligands stimulate NSC survival and proliferation in a dosage-dependent manner in vitro. NogoR1 and PirB are low and high affinity Nogo receptors, respectively and are responsible for the effects of Nogo ligands on NSC behavior. Inhibition of autocrine-paracrine Nogo signaling blocks NSC survival and proliferation. In NSCs, NogoR1 functions through Rho whereas PirB uses Shp1/2 signaling pathways to control NSC behavior. Taken together, this work suggests that Nogo signaling is an important pathway for survival of NSCs.
Collapse
Affiliation(s)
- Srinivas Ramasamy
- Institute of Medical Biology, 8A Biomedical Grove, #05-37 Immunos, Singapore
| | | | | | | | | | | |
Collapse
|
33
|
Thiede-Stan NK, Tews B, Albrecht D, Ristic Z, Ewers H, Schwab ME. Tetraspanin-3 is an organizer of the multi-subunit Nogo-A signaling complex. J Cell Sci 2015; 128:3583-96. [DOI: 10.1242/jcs.167981] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 08/17/2015] [Indexed: 01/01/2023] Open
Abstract
To ensure precision and specificity of ligand – receptor induced signaling, co-receptors and modulatory factors play important roles. The membrane bound ligand Nogo-A induces inhibition of neurite outgrowth, cell spreading, adhesion and migration via multi-subunit receptor complexes. Here, we identified the 4-transmembrane-spanning protein tetraspanin-3 (TSPAN3) as a new modulatory co-receptor for the Nogo-A inhibitory domain Nogo-A-Δ20. Single-molecule-tracking showed that TSPAN3 molecules in the cell membrane reacted with elevated mobility to Nogo-A binding, followed by association with the signal transducing Nogo-A receptor sphingosine-1-phosphate receptor 2 (S1PR2). Subsequently, TSPAN3 was co-internalized as part of the Nogo-A ligand – receptor complex into early endosomes, where it subsequently separated from Nogo-A and S1PR2 to be recycled to the cell surface. The functional importance of the Nogo-A – TSPAN3 interaction is shown by the fact that knockdown of TSPAN3 strongly reduced the Nogo-A-induced S1PR2 clustering, RhoA activation and cell spreading and neurite outgrowth inhibition. In addition to the modulatory functions of TSPAN3 on Nogo-A-S1PR2 signaling, these results illustrate the very dynamic spatiotemporal reorganizations of membrane proteins during ligand-induced receptor complex organizations.
Collapse
Affiliation(s)
- Nina K. Thiede-Stan
- Brain Research Institute, University of Zurich and Dept. of Health Sciences & Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Björn Tews
- Brain Research Institute, University of Zurich and Dept. of Health Sciences & Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - David Albrecht
- Institute of Biochemistry and Laboratory of Physical Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Zorica Ristic
- Brain Research Institute, University of Zurich and Dept. of Health Sciences & Technology, ETH Zurich, 8057 Zurich, Switzerland
| | - Helge Ewers
- Institute of Biochemistry and Laboratory of Physical Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich and Dept. of Health Sciences & Technology, ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
34
|
Boda E, Di Maria S, Rosa P, Taylor V, Abbracchio MP, Buffo A. Early phenotypic asymmetry of sister oligodendrocyte progenitor cells after mitosis and its modulation by aging and extrinsic factors. Glia 2014; 63:271-86. [PMID: 25213035 DOI: 10.1002/glia.22750] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/22/2014] [Indexed: 01/26/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs) persist in the adult central nervous system and guarantee oligodendrocyte turnover throughout life. It remains obscure how OPCs avoid exhaustion during adulthood. Similar to stem cells, OPCs could self-maintain by undergoing asymmetric divisions generating a mixed progeny either keeping a progenitor phenotype or proceeding to differentiation. To address this issue, we examined the distribution of stage-specific markers in sister OPCs during mitosis and later after cell birth, and assessed its correlation with distinct short-term fates. In both the adult and juvenile cerebral cortex a fraction of dividing OPCs gives rise to sister cells with diverse immunophenotypic profiles and short-term behaviors. Such heterogeneity appears as cells exit cytokinesis, but does not derive from the asymmetric segregation of molecules such as NG2 or PDGFRa expressed in the mother cell. Rather, rapid downregulation of OPC markers and upregulation of molecules associated with lineage progression contributes to generate early sister OPC asymmetry. Analyses during aging and upon exposure to physiological (i.e., increased motor activity) and pathological (i.e., trauma or demyelination) stimuli showed that both intrinsic and environmental factors contribute to determine the fraction of symmetric and asymmetric OPC pairs and the phenotype of the OPC progeny as soon as cells exit mitosis.
Collapse
Affiliation(s)
- Enrica Boda
- Department of Neuroscience, Neuroscience Institute Cavalieri Ottolenghi (NICO), Università degli Studi di Torino, Regione Gonzole, 10-10043, Orbassano (Turin), Italy
| | | | | | | | | | | |
Collapse
|
35
|
Neutralization of Nogo-A enhances synaptic plasticity in the rodent motor cortex and improves motor learning in vivo. J Neurosci 2014; 34:8685-98. [PMID: 24966370 DOI: 10.1523/jneurosci.3817-13.2014] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The membrane protein Nogo-A is known as an inhibitor of axonal outgrowth and regeneration in the CNS. However, its physiological functions in the normal adult CNS remain incompletely understood. Here, we investigated the role of Nogo-A in cortical synaptic plasticity and motor learning in the uninjured adult rodent motor cortex. Nogo-A and its receptor NgR1 are present at cortical synapses. Acute treatment of slices with function-blocking antibodies (Abs) against Nogo-A or against NgR1 increased long-term potentiation (LTP) induced by stimulation of layer 2/3 horizontal fibers. Furthermore, anti-Nogo-A Ab treatment increased LTP saturation levels, whereas long-term depression remained unchanged, thus leading to an enlarged synaptic modification range. In vivo, intrathecal application of Nogo-A-blocking Abs resulted in a higher dendritic spine density at cortical pyramidal neurons due to an increase in spine formation as revealed by in vivo two-photon microscopy. To investigate whether these changes in synaptic plasticity correlate with motor learning, we trained rats to learn a skilled forelimb-reaching task while receiving anti-Nogo-A Abs. Learning of this cortically controlled precision movement was improved upon anti-Nogo-A Ab treatment. Our results identify Nogo-A as an influential molecular modulator of synaptic plasticity and as a regulator for learning of skilled movements in the motor cortex.
Collapse
|
36
|
Roll L, Faissner A. Influence of the extracellular matrix on endogenous and transplanted stem cells after brain damage. Front Cell Neurosci 2014; 8:219. [PMID: 25191223 PMCID: PMC4137450 DOI: 10.3389/fncel.2014.00219] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/18/2014] [Indexed: 01/07/2023] Open
Abstract
The limited regeneration capacity of the adult central nervous system (CNS) requires strategies to improve recovery of patients. In this context, the interaction of endogenous as well as transplanted stem cells with their environment is crucial. An understanding of the molecular mechanisms could help to improve regeneration by targeted manipulation. In the course of reactive gliosis, astrocytes upregulate Glial fibrillary acidic protein (GFAP) and start, in many cases, to proliferate. Beside GFAP, subpopulations of these astroglial cells coexpress neural progenitor markers like Nestin. Although cells express these markers, the proportion of cells that eventually give rise to neurons is limited in many cases in vivo compared to the situation in vitro. In the first section, we present the characteristics of endogenous progenitor-like cells and discuss the differences in their neurogenic potential in vitro and in vivo. As the environment plays an important role for survival, proliferation, migration, and other processes, the second section of the review describes changes in the extracellular matrix (ECM), a complex network that contains numerous signaling molecules. It appears that signals in the damaged CNS lead to an activation and de-differentiation of astrocytes, but do not effectively promote neuronal differentiation of these cells. Factors that influence stem cells during development are upregulated in the damaged brain as part of an environment resembling a stem cell niche. We give a general description of the ECM composition, with focus on stem cell-associated factors like the glycoprotein Tenascin-C (TN-C). Stem cell transplantation is considered as potential treatment strategy. Interaction of transplanted stem cells with the host environment is critical for the outcome of stem cell-based therapies. Possible mechanisms involving the ECM by which transplanted stem cells might improve recovery are discussed in the last section.
Collapse
Affiliation(s)
- Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany ; International Graduate School of Neuroscience, Ruhr-University Bochum Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum Bochum, Germany ; International Graduate School of Neuroscience, Ruhr-University Bochum Bochum, Germany
| |
Collapse
|
37
|
Ota H, Hikita T, Sawada M, Nishioka T, Matsumoto M, Komura M, Ohno A, Kamiya Y, Miyamoto T, Asai N, Enomoto A, Takahashi M, Kaibuchi K, Sobue K, Sawamoto K. Speed control for neuronal migration in the postnatal brain by Gmip-mediated local inactivation of RhoA. Nat Commun 2014; 5:4532. [DOI: 10.1038/ncomms5532] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/26/2014] [Indexed: 01/04/2023] Open
|
38
|
Pinzón-Olejua A, Welte C, Abdesselem H, Málaga-Trillo E, Stuermer CA. Essential roles of zebrafish rtn4/Nogo paralogues in embryonic development. Neural Dev 2014; 9:8. [PMID: 24755266 PMCID: PMC4113184 DOI: 10.1186/1749-8104-9-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 03/25/2014] [Indexed: 01/08/2023] Open
Abstract
Background As a consequence of gene/genome duplication, the RTN4/Nogo gene has two counterparts in zebrafish: rtn4a and rtn4b. The shared presence of four specific amino acid motifs—M1 to M4—in the N-terminal region of mammalian RTN4, and zebrafish Rtn4b suggests that Rtn4b is the closest homologue of mammalian Nogo-A. Results To explore their combined roles in zebrafish development, we characterized the expression patterns of rtn4a and rtn4b in a comparative manner and performed morpholino-mediated knockdowns. Although both genes were coexpressed in the neural tube and developing brain at early stages, they progressively acquired distinct expression domains such as the spinal cord (rtn4b) and somites (rtn4a). Downregulation of rtn4a and rtn4b caused severe brain abnormalities, with rtn4b knockdown severely affecting the spinal cord and leading to immobility. In addition, the retinotectal projection was severely affected in both morphants, as the retina and optic tectum appeared smaller and only few retinal axons reached the abnormally reduced tectal neuropil. The neuronal defects were more persistent in rtn4b morphants. Moreover, the latter often lacked pectoral fins and lower jaws and had malformed branchial arches. Notably, these defects led to larval death in rtn4b, but not in rtn4a morphants. Conclusions In contrast to mammalian Nogo-A, its zebrafish homologues, rtn4a and particularly rtn4b, are essential for embryonic development and patterning of the nervous system.
Collapse
Affiliation(s)
| | | | | | - Edward Málaga-Trillo
- Department of Biology, University of Konstanz, Universitätsstrasse 10, 78476 Konstanz, Germany.
| | | |
Collapse
|
39
|
Petrasek T, Prokopova I, Sladek M, Weissova K, Vojtechova I, Bahnik S, Zemanova A, Schönig K, Berger S, Tews B, Bartsch D, Schwab ME, Sumova A, Stuchlik A. Nogo-A-deficient Transgenic Rats Show Deficits in Higher Cognitive Functions, Decreased Anxiety, and Altered Circadian Activity Patterns. Front Behav Neurosci 2014; 8:90. [PMID: 24672453 PMCID: PMC3957197 DOI: 10.3389/fnbeh.2014.00090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/02/2014] [Indexed: 11/19/2022] Open
Abstract
Decreased levels of Nogo-A-dependent signaling have been shown to affect behavior and cognitive functions. In Nogo-A knockout and knockdown laboratory rodents, behavioral alterations were observed, possibly corresponding with human neuropsychiatric diseases of neurodevelopmental origin, particularly schizophrenia. This study offers further insight into behavioral manifestations of Nogo-A knockdown in laboratory rats, focusing on spatial and non-spatial cognition, anxiety levels, circadian rhythmicity, and activity patterns. Demonstrated is an impairment of cognitive functions and behavioral flexibility in a spatial active avoidance task, while non-spatial memory in a step-through avoidance task was spared. No signs of anhedonia, typical for schizophrenic patients, were observed in the animals. Some measures indicated lower anxiety levels in the Nogo-A-deficient group. Circadian rhythmicity in locomotor activity was preserved in the Nogo-A knockout rats and their circadian period (tau) did not differ from controls. However, daily activity patterns were slightly altered in the knockdown animals. We conclude that a reduction of Nogo-A levels induces changes in CNS development, manifested as subtle alterations in cognitive functions, emotionality, and activity patterns.
Collapse
Affiliation(s)
- Tomas Petrasek
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic ; First Faculty of Medicine, Charles University in Prague , Prague , Czech Republic
| | - Iva Prokopova
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Martin Sladek
- Department of Neurohumoral Regulations, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Kamila Weissova
- Department of Neurohumoral Regulations, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Iveta Vojtechova
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Stepan Bahnik
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic ; Social Psychology, Department of Psychology II, University of Würzburg , Würzburg , Germany
| | - Anna Zemanova
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Kai Schönig
- Department of Molecular Biology, Central Institute of Mental Health , Mannheim , Germany
| | - Stefan Berger
- Department of Molecular Biology, Central Institute of Mental Health , Mannheim , Germany
| | - Björn Tews
- Brain Research Institute, University of Zurich , Zurich , Switzerland ; Neurosciences, Department of Biology, Swiss Federal Institute of Technology Zurich , Zurich , Switzerland ; Division of Molecular Mechanisms of Tumor Invasion, German Cancer Research Center , Heidelberg , Germany
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health , Mannheim , Germany
| | - Martin E Schwab
- Brain Research Institute, University of Zurich , Zurich , Switzerland ; Neurosciences, Department of Biology, Swiss Federal Institute of Technology Zurich , Zurich , Switzerland
| | - Alena Sumova
- Department of Neurohumoral Regulations, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Ales Stuchlik
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| |
Collapse
|
40
|
Canazza A, Minati L, Boffano C, Parati E, Binks S. Experimental models of brain ischemia: a review of techniques, magnetic resonance imaging, and investigational cell-based therapies. Front Neurol 2014; 5:19. [PMID: 24600434 PMCID: PMC3928567 DOI: 10.3389/fneur.2014.00019] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 02/06/2014] [Indexed: 01/17/2023] Open
Abstract
Stroke continues to be a significant cause of death and disability worldwide. Although major advances have been made in the past decades in prevention, treatment, and rehabilitation, enormous challenges remain in the way of translating new therapeutic approaches from bench to bedside. Thrombolysis, while routinely used for ischemic stroke, is only a viable option within a narrow time window. Recently, progress in stem cell biology has opened up avenues to therapeutic strategies aimed at supporting and replacing neural cells in infarcted areas. Realistic experimental animal models are crucial to understand the mechanisms of neuronal survival following ischemic brain injury and to develop therapeutic interventions. Current studies on experimental stroke therapies evaluate the efficiency of neuroprotective agents and cell-based approaches using primarily rodent models of permanent or transient focal cerebral ischemia. In parallel, advancements in imaging techniques permit better mapping of the spatial-temporal evolution of the lesioned cortex and its functional responses. This review provides a condensed conceptual review of the state of the art of this field, from models and magnetic resonance imaging techniques through to stem cell therapies.
Collapse
Affiliation(s)
- Alessandra Canazza
- Cerebrovascular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan , Italy
| | - Ludovico Minati
- Scientific Department, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan , Italy ; Brighton and Sussex Medical School , Brighton , UK
| | - Carlo Boffano
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan , Italy
| | - Eugenio Parati
- Cerebrovascular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan , Italy
| | - Sophie Binks
- Brighton and Sussex Medical School , Brighton , UK ; Brighton and Sussex University Hospitals NHS Trust , Brighton , UK
| |
Collapse
|
41
|
Lalli G. Extracellular Signals Controlling Neuroblast Migration in the Postnatal Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 800:149-80. [DOI: 10.1007/978-94-007-7687-6_9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Boccazzi M, Rolando C, Abbracchio MP, Buffo A, Ceruti S. Purines regulate adult brain subventricular zone cell functions: contribution of reactive astrocytes. Glia 2013; 62:428-39. [PMID: 24382645 DOI: 10.1002/glia.22614] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/19/2013] [Accepted: 11/21/2013] [Indexed: 12/21/2022]
Abstract
Brain injuries modulate activation of neural stem cells (NSCs) in the adult brain. In pathological conditions, the concentrations of extracellular nucleotides (eNTs) raise several folds, contribute to reactive gliosis, and possibly directly affect subventricular zone (SVZ) cell functioning. Among eNTs and derived metabolites, the P2Y1 receptor agonist ADP strongly promotes astrogliosis and might also influence SVZ progenitor activity. Here, we tested the ability of the stable P2Y1 agonist adenosine 5'-O-(2-thiodiphosphate) (ADPβS) to control adult NSC functions both in vitro and in vivo, with a focus on the possible effects exerted by reactive astrocytes. In the absence of growth factors, ADPβS promoted proliferation and differentiation of SVZ progenitors. Moreover, ADPβS-activated astrocytes markedly changed the pattern of released cytokines and chemokines, and strongly modulated neurosphere-forming capacity of SVZ progenitors. Notably, a significant enhancement in proliferation was observed when SVZ cells, initially grown in the supernatant of astrocytes exposed to ADPβS, were shifted to normal medium. In vivo, ADPβS administration in the lateral ventricle of adult mice by osmotic minipumps caused diffused reactive astrogliosis, and a strong response of SVZ progenitors. Indeed, proliferation of glial fibrillary acidic protein-positive NSCs increased and led to a significant expansion of SVZ transit-amplifying progenitors and neuroblasts. Lineage tracing experiments performed in the GLAST::CreERT2;Rosa-YFP transgenic mice further demonstrated that ADPβS promoted proliferation of glutamate/aspartate transporter-positive progenitors and sustained their progression toward the generation of rapidly dividing progenitors. Altogether, our results show that the purinergic system crucially affects SVZ progenitor activities both directly and through the involvement of reactive astrocytes.
Collapse
Affiliation(s)
- Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | |
Collapse
|
43
|
Schmandke A, Mosberger AC, Schmandke A, Celen Z, Schwab ME. The neurite growth inhibitory protein Nogo-A has diverse roles in adhesion and migration. Cell Adh Migr 2013; 7:451-4. [PMID: 24401759 DOI: 10.4161/cam.27164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Andre Schmandke
- Brain Research Institute; University of Zurich and Department of Health Sciences and Technology; ETH Zurich, Winterthurerstrasse 190; Zurich, Switzerland
| | - Alice C Mosberger
- Brain Research Institute; University of Zurich and Department of Health Sciences and Technology; ETH Zurich, Winterthurerstrasse 190; Zurich, Switzerland
| | - Antonio Schmandke
- Brain Research Institute; University of Zurich and Department of Health Sciences and Technology; ETH Zurich, Winterthurerstrasse 190; Zurich, Switzerland
| | - Zeliha Celen
- Brain Research Institute; University of Zurich and Department of Health Sciences and Technology; ETH Zurich, Winterthurerstrasse 190; Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute; University of Zurich and Department of Health Sciences and Technology; ETH Zurich, Winterthurerstrasse 190; Zurich, Switzerland
| |
Collapse
|
44
|
Schmandke A, Schmandke A, Pietro MA, Schwab ME. An open source based high content screening method for cell biology laboratories investigating cell spreading and adhesion. PLoS One 2013; 8:e78212. [PMID: 24205161 PMCID: PMC3804740 DOI: 10.1371/journal.pone.0078212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/18/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Adhesion dependent mechanisms are increasingly recognized to be important for a wide range of biological processes, diseases and therapeutics. This has led to a rising demand of pharmaceutical modulators. However, most currently available adhesion assays are time consuming and/or lack sensitivity and reproducibility or depend on specialized and expensive equipment often only available at screening facilities. Thus, rapid and economical high-content screening approaches are urgently needed. RESULTS We established a fully open source high-content screening method for identifying modulators of adhesion. We successfully used this method to detect small molecules that are able to influence cell adhesion and cell spreading of Swiss-3T3 fibroblasts in general and/or specifically counteract Nogo-A-Δ20-induced inhibition of adhesion and cell spreading. The tricyclic anti-depressant clomipramine hydrochloride was shown to not only inhibit Nogo-A-Δ20-induced cell spreading inhibition in 3T3 fibroblasts but also to promote growth and counteract neurite outgrowth inhibition in highly purified primary neurons isolated from rat cerebellum. CONCLUSIONS We have developed and validated a high content screening approach that can be used in any ordinarily equipped cell biology laboratory employing exclusively freely available open-source software in order to find novel modulators of adhesion and cell spreading. The versatility and adjustability of the whole screening method will enable not only centers specialized in high-throughput screens but most importantly also labs not routinely employing screens in their daily work routine to investigate the effects of a wide range of different compounds or siRNAs on adhesion and adhesion-modulating molecules.
Collapse
Affiliation(s)
- Andre Schmandke
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Antonio Schmandke
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Maurianne A. Pietro
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
45
|
Persson A, Lindberg OR, Kuhn HG. Radixin inhibition decreases adult neural progenitor cell migration and proliferation in vitro and in vivo. Front Cell Neurosci 2013; 7:161. [PMID: 24065889 PMCID: PMC3781578 DOI: 10.3389/fncel.2013.00161] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/03/2013] [Indexed: 11/13/2022] Open
Abstract
Neuronal progenitors capable of long distance migration are produced throughout life in the subventricular zone (SVZ). Migration from the SVZ is carried out along a well-defined pathway called the rostral migratory stream (RMS). Our recent finding of the specific expression of the cytoskeleton linker protein radixin in neuroblasts suggests a functional role for radixin in RMS migration. The ezrin-radixin-moesin (ERM) family of proteins is capable of regulating migration through interaction with the actin cytoskeleton and transmembrane proteins. The ERM proteins are differentially expressed in the RMS with radixin and moesin localized to neuroblasts, and ezrin expression confined to astrocytes of the glial tubes. Here, we inhibited radixin function using the quinocarmycin analog DX52-1 which resulted in reduced neuroblast migration in vitro, while glial migration remained unaltered. Furthermore, the morphology of neuroblasts was distorted resulting in a rounded shape with no or short polysialylated neural cell adhesion molecule positive processes. Intracerebroventricular infusion of the radixin inhibitor resulted in accumulation of neuroblasts in the anterior SVZ. Neuroblast chains were short and intermittently interrupted in the SVZ and considerably disorganized in the RMS. Moreover, we studied the proliferation activity in the RMS after radixin inhibition, since concentrated radixin expression has been demonstrated in the cleavage furrow of dividing cells, which indicates a role of radixin in cell division. Radixin inhibition decreased neuroblast proliferation, whereas the proliferation of other cells in the RMS was not affected. Our results demonstrate a significant role for radixin in neuroblast proliferation and migration.
Collapse
Affiliation(s)
- Asa Persson
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg Gothenburg, Sweden
| | | | | |
Collapse
|
46
|
Wang J, Li X, Cheng H, Wang K, Lu W, Wen T. Overexpression of Rho-GDP-dissociation inhibitor-γ inhibits migration of neural stem cells. J Neurosci Res 2013; 91:1394-401. [PMID: 23996536 DOI: 10.1002/jnr.23261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/03/2013] [Accepted: 05/21/2013] [Indexed: 12/11/2022]
Abstract
Neural stem cell (NSC) migration relies heavily on the regulation of actin and microtubule cytoskeletons by Rho GTPases, which are critical regulators of key steps during NSC migration. However, the migration mechanism remains unclear. Rho-GDP-dissociation inhibitor-γ (Rho-GDIγ) was identified as an important downregulator of the Rho family of GTPases, because of its ability to prevent nucleotide exchange and thus membrane association. This study investigates the role of Rho-GDIγ in neural stem cells migration. Our results indicate that the overexpression of Rho-GDIγ maintains NSCs in the stem cell state, meanwhile preventing NSC migration through inhibition of Rac1 expression, one of the Rho-family GTPases. This study provides the basis for further study of the molecular mechanism of NSC migration.
Collapse
Affiliation(s)
- Jiao Wang
- Laboratory of Molecular Neural Biology, Institute of Systems Biology, School of Life Sciences, Shanghai University, Shanghai, China; School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
47
|
Kempf A, Schwab ME. Nogo-A Represses Anatomical and Synaptic Plasticity in the Central Nervous System. Physiology (Bethesda) 2013; 28:151-63. [DOI: 10.1152/physiol.00052.2012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Nogo-A was initially discovered as a myelin-associated growth inhibitory protein limiting axonal regeneration after central nervous system (CNS) injury. This review summarizes current knowledge on how myelin and neuronal Nogo-A and its receptors exert physiological functions ranging from the regulation of growth suppression to synaptic plasticity in the developing and adult intact CNS.
Collapse
Affiliation(s)
- Anissa Kempf
- Brain Research Institute, University of Zurich, and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich, and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Li T, Pan YW, Wang W, Abel G, Zou J, Xu L, Storm DR, Xia Z. Targeted deletion of the ERK5 MAP kinase impairs neuronal differentiation, migration, and survival during adult neurogenesis in the olfactory bulb. PLoS One 2013; 8:e61948. [PMID: 23630619 PMCID: PMC3632513 DOI: 10.1371/journal.pone.0061948] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 03/15/2013] [Indexed: 01/13/2023] Open
Abstract
Recent studies have led to the exciting idea that adult-born neurons in the olfactory bulb (OB) may be critical for complex forms of olfactory behavior in mice. However, signaling mechanisms regulating adult OB neurogenesis are not well defined. We recently reported that extracellular signal-regulated kinase (ERK) 5, a MAP kinase, is specifically expressed in neurogenic regions within the adult brain. This pattern of expression suggests a role for ERK5 in the regulation of adult OB neurogenesis. Indeed, we previously reported that conditional deletion of erk5 in adult neurogenic regions impairs several forms of olfactory behavior in mice. Thus, it is important to understand how ERK5 regulates adult neurogenesis in the OB. Here we present evidence that shRNA suppression of ERK5 in adult neural stem/progenitor cells isolated from the subventricular zone (SVZ) reduces neurogenesis in culture. By contrast, ectopic activation of endogenous ERK5 signaling via expression of constitutive active MEK5, an upstream activating kinase for ERK5, stimulates neurogenesis. Furthermore, inducible and conditional deletion of erk5 specifically in the neurogenic regions of the adult mouse brain interferes with cell cycle exit of neuroblasts, impairs chain migration along the rostral migratory stream and radial migration into the OB. It also inhibits neuronal differentiation and survival. These data suggest that ERK5 regulates multiple aspects of adult OB neurogenesis and provide new insights concerning signaling mechanisms governing adult neurogenesis in the SVZ-OB axis.
Collapse
Affiliation(s)
- Tan Li
- Toxicology Program in the Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
- Department of Biochemistry and Genetics, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yung-Wei Pan
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, Washington, United States of America
| | - Wenbin Wang
- Toxicology Program in the Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Glen Abel
- Toxicology Program in the Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Junhui Zou
- Toxicology Program in the Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Lihong Xu
- Department of Biochemistry and Genetics, Zhejiang University, Hangzhou, Zhejiang, China
| | - Daniel R. Storm
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Zhengui Xia
- Toxicology Program in the Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
49
|
Abstract
Over the past decade, there has been substantial interest in the role of the integral myelin protein, Nogo-A, from fundamental neurobiological to clinical perspectives. It is now a well-known inhibitor of neurite outgrowth through its cognate receptor, Nogo receptor 1 (NgR1). Nogo-A can only signal through NgR1 upon heteromeric collaboration with p75(NTR), TROY, and LINGO-1 to induce axonal retraction. Both Nogo-A and NgR1 are expressed in multiple sclerosis (MS) lesions, suggesting that Nogo signaling may play a pivotal role in disease progression. There are several approaches targeting Nogo signaling in animal models of MS, and these therapeutic effects are currently in debate. One of the points of contention arises from the localization of the aforementioned signaling molecules, considering that MS and its animal models of disease are governed by inflammatory infiltration of the central nervous system. Furthermore, an impressive list of ligands for NgR1 continues to be compiled, possibly leading to disparities in the results obtained from the various animal models. In this review, we systematically dissect the complexities of Nogo signaling, which may be relevant in the future directions of neuroprotective therapies for MS.
Collapse
Affiliation(s)
- Jae Young Lee
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|