1
|
Del Casale A, Modesti MN, Gentile G, Guariglia C, Ferracuti S, Simmaco M, Borro M. Is the Hedgehog Pathway Involved in the Pathophysiology of Schizophrenia? A Systematic Review of Current Evidence of Neural Molecular Correlates and Perspectives on Drug Development. Curr Issues Mol Biol 2024; 46:5322-5336. [PMID: 38920990 PMCID: PMC11202070 DOI: 10.3390/cimb46060318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Among the pathophysiological correlates of schizophrenia, recent research suggests a potential role for the Hedgehog (Hh) signalling pathway, which has been traditionally studied in embryonic development and oncology. Its dysregulation may impact brain homeostasis, neuroplasticity, and potential involvement in neural processes. This systematic review provides an overview of the involvement of Hh signalling in the pathophysiology of schizophrenia and antipsychotic responses. We searched the PubMed and Scopus databases to identify peer-reviewed scientific studies focusing on Hh and schizophrenia, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement, finally including eight studies, including three articles focused on patients with schizophrenia, two animal models of schizophrenia, two animal embryo studies, and one cellular differentiation study. The Hh pathway is crucial in the development of midbrain dopaminergic neurons, neuroplasticity mechanisms, regulating astrocyte phenotype and function, brain-derived neurotrophic factor expression, brain glutamatergic neural transmission, and responses to antipsychotics. Overall, results indicate an involvement of Hh in the pathophysiology of schizophrenia and antipsychotic responses, although an exiguity of studies characterises the literature. The heterogeneity between animal and human studies is another main limitation. Further research can lead to better comprehension and the development of novel personalised drug treatments and therapeutic interventions.
Collapse
Affiliation(s)
- Antonio Del Casale
- Department of Dynamic and Clinical Psychology and Health Studies, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy;
- Unit of Psychiatry, Emergency and Admissions Department, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Martina Nicole Modesti
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Unit of Psychiatry, Mental Health Department, Santissimo Gonfalone Hospital, Local Health Service Roma 5, Monterotondo, 00015 Rome, Italy
| | - Giovanna Gentile
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Cecilia Guariglia
- Department of Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome, 00185 Rome, Italy
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, Scientific Institute for Research, Hospitalization and Healthcare Fondazione Santa Lucia, 00179 Rome, Italy
| | - Stefano Ferracuti
- Department of Human Neuroscience, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00185 Rome, Italy;
- Unit of Risk Management, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Maurizio Simmaco
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Marina Borro
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
- Unit of Laboratory and Advanced Molecular Diagnostics, Sant’Andrea University Hospital, 00189 Rome, Italy
| |
Collapse
|
2
|
Russo M, Pellegrino G, Faure H, Tirou L, Sharif A, Ruat M. Characterization of Sonic Hedgehog transcripts in the adult mouse brain: co-expression with neuronal and oligodendroglial markers. Brain Struct Funct 2024; 229:705-727. [PMID: 38329543 PMCID: PMC10978748 DOI: 10.1007/s00429-023-02756-2] [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: 08/31/2023] [Accepted: 12/29/2023] [Indexed: 02/09/2024]
Abstract
In the adult mammalian brain, astrocytes are proposed to be the major Sonic Hedgehog (Shh)-responsive cells. However, the sources of the Shh molecule mediating activation of the pathway are still poorly characterized. The present work investigates the distribution and phenotype of cells expressing Shh mRNA in the adult mouse brain. Using single-molecule fluorescent in situ hybridization (smfISH), we report much broader expression of Shh transcripts in almost all brain regions than originally reported. We identify Shh mRNA in HuC/D+ neuronal populations, including GABAergic (glutamic acid decarboxylase 67, Gad67), cholinergic (choline acetyltransferase, ChAT), dopaminergic (tyrosine hydroxylase, TH), nitrergic (neuronal nitric oxide synthase, nNOS), and in a small population of oligodendroglial cells expressing Sox10 and Olig2 mRNA transcription factors. Further analysis of Shh mRNA in cerebral cortical and hypothalamic neurons suggests that Shh is also expressed by glutamatergic neurons. Interestingly, we did not observe substantial Desert Hedgehog and Indian Hedgehog mRNA signals, nor Shh signals in S100β+ astrocytes and Iba1+ microglial cells. Collectively, the present work provides the most robust central map of Shh-expressing cells to date and underscores the importance of nitrergic neurons in regulating Shh availability to brain cells. Thus, our study provides a framework for future experiments aimed at better understanding of the functions of Shh signaling in the brain in normal and pathological states, and the characterization of novel regulatory mechanisms of the signaling pathway.
Collapse
Affiliation(s)
- Mariagiovanna Russo
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Giuliana Pellegrino
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Hélène Faure
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Linda Tirou
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France
| | - Martial Ruat
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France.
| |
Collapse
|
3
|
Duzan A, Reinken D, McGomery TL, Ferencz NM, Plummer JM, Basti MM. Endocannabinoids are potential inhibitors of glioblastoma multiforme proliferation. JOURNAL OF INTEGRATIVE MEDICINE 2023; 21:120-129. [PMID: 36805391 DOI: 10.1016/j.joim.2023.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/14/2022] [Indexed: 02/04/2023]
Abstract
Globally, it is evident that glioblastoma multiforme (GBM) is an aggressive malignant cancer with a high mortality rate and no effective treatment options. Glioblastoma is classified as the stage-four progression of a glioma tumor, and its diagnosis results in a shortened life expectancy. Treatment options for GBM include chemotherapy, immunotherapy, surgical intervention, and conventional pharmacotherapy; however, at best, they extend the patient's life by a maximum of 5 years. GBMs are considered incurable due to their high recurrence rate, despite various aggressive therapeutic approaches which can have many serious adverse effects. Ceramides, classified as endocannabinoids, offer a promising novel therapeutic approach for GBM. Endocannabinoids may enhance the apoptosis of GBM cells but have no effect on normal healthy neural cells. Cannabinoids promote atypical protein kinase C, deactivate fatty acid amide hydrolase enzymes, and activate transient receptor potential vanilloid 1 (TRPV1) and TRPV2 to induce pro-apoptotic signaling pathways without increasing endogenous cannabinoids. In previous in vivo studies, endocannabinoids, chemically classified as amide formations of oleic and palmitic acids, have been shown to increase the pro-apoptotic activity of human cancer cells and inhibit cell migration and angiogenesis. This review focuses on the biological synthesis and pharmacology of endogenous cannabinoids for the enhancement of cancer cell apoptosis, which have potential as a novel therapy for GBM. Please cite this article as: Duzan A, Reinken D, McGomery TL, Ferencz N, Plummer JM, Basti MM. Endocannabinoids are potential inhibitors of glioblastoma multiforme proliferation. J Integr Med. 2023; Epub ahead of print.
Collapse
Affiliation(s)
- Ashraf Duzan
- School of Pharmacy, Wingate University, Wingate, NC 28174, USA; Applied Science and Technology Department, North Carolina State University of Agriculture and Technology, Greensboro, NC 27411, USA.
| | - Desiree Reinken
- College of Nursing, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | | | - Jacob M Plummer
- Collage of Arts and Science, Department of Chemistry and Physics, Wingate University, Wingate, NC 28174, USA
| | - Mufeed M Basti
- Applied Science and Technology Department, North Carolina State University of Agriculture and Technology, Greensboro, NC 27411, USA.
| |
Collapse
|
4
|
Douceau S, Deutsch Guerrero T, Ferent J. Establishing Hedgehog Gradients during Neural Development. Cells 2023; 12:225. [PMID: 36672161 PMCID: PMC9856818 DOI: 10.3390/cells12020225] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/07/2023] Open
Abstract
A morphogen is a signaling molecule that induces specific cellular responses depending on its local concentration. The concept of morphogenic gradients has been a central paradigm of developmental biology for decades. Sonic Hedgehog (Shh) is one of the most important morphogens that displays pleiotropic functions during embryonic development, ranging from neuronal patterning to axon guidance. It is commonly accepted that Shh is distributed in a gradient in several tissues from different origins during development; however, how these gradients are formed and maintained at the cellular and molecular levels is still the center of a great deal of research. In this review, we first explored all of the different sources of Shh during the development of the nervous system. Then, we detailed how these sources can distribute Shh in the surrounding tissues via a variety of mechanisms. Finally, we addressed how disrupting Shh distribution and gradients can induce severe neurodevelopmental disorders and cancers. Although the concept of gradient has been central in the field of neurodevelopment since the fifties, we also describe how contemporary leading-edge techniques, such as organoids, can revisit this classical model.
Collapse
Affiliation(s)
- Sara Douceau
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| | - Tanya Deutsch Guerrero
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| | - Julien Ferent
- INSERM UMR-S 1270, F-75005 Paris, France
- Institut du Fer à Moulin, INSERM, Sorbonne Univeristy, F-75005 Paris, France
| |
Collapse
|
5
|
Zhang C, Xue P, Zhang H, Tan C, Zhao S, Li X, Sun L, Zheng H, Wang J, Zhang B, Lang W. Gut brain interaction theory reveals gut microbiota mediated neurogenesis and traditional Chinese medicine research strategies. Front Cell Infect Microbiol 2022; 12:1072341. [PMID: 36569198 PMCID: PMC9772886 DOI: 10.3389/fcimb.2022.1072341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022] Open
Abstract
Adult neurogenesis is the process of differentiation of neural stem cells (NSCs) into neurons and glial cells in certain areas of the adult brain. Defects in neurogenesis can lead to neurodegenerative diseases, mental disorders, and other maladies. This process is directionally regulated by transcription factors, the Wnt and Notch pathway, the extracellular matrix, and various growth factors. External factors like stress, physical exercise, diet, medications, etc., affect neurogenesis and the gut microbiota. The gut microbiota may affect NSCs through vagal, immune and chemical pathways, and other pathways. Traditional Chinese medicine (TCM) has been proven to affect NSCs proliferation and differentiation and can regulate the abundance and metabolites produced by intestinal microorganisms. However, the underlying mechanisms by which these factors regulate neurogenesis through the gut microbiota are not fully understood. In this review, we describe the recent evidence on the role of the gut microbiota in neurogenesis. Moreover, we hypothesize on the characteristics of the microbiota-gut-brain axis based on bacterial phyla, including microbiota's metabolites, and neuronal and immune pathways while providing an outlook on TCM's potential effects on adult neurogenesis by regulating gut microbiota.
Collapse
Affiliation(s)
- Chenxi Zhang
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Peng Xue
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Haiyan Zhang
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Chenxi Tan
- Department of Infection Control, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Shiyao Zhao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Xudong Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lihui Sun
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Huihui Zheng
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Jun Wang
- The Academic Affairs Office, Qiqihar Medical University, Qiqihar, China
| | - Baoling Zhang
- Department of Operating Room, Qiqihar First Hospital, Qiqihar, China
| | - Weiya Lang
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China,*Correspondence: Weiya Lang,
| |
Collapse
|
6
|
Rava A, La Rosa P, Palladino G, Dragotto J, Totaro A, Tiberi J, Canterini S, Oddi S, Fiorenza MT. The appearance of phagocytic microglia in the postnatal brain of Niemann Pick type C mice is developmentally regulated and underscores shortfalls in fine odor discrimination. J Cell Physiol 2022; 237:4563-4579. [PMID: 36322609 PMCID: PMC7613956 DOI: 10.1002/jcp.30909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
The loss of NPC1 or NPC2 function results in cholesterol and sphingolipid dyshomeostasis that impairs developmental trajectories, predisposing the postnatal brain to the appearance of pathological signs, including progressive and stereotyped Purkinje cell loss and microgliosis. Despite increasing evidence reporting the activation of pro-inflammatory microglia as a cardinal event of NPC1 disease progression at symptomatic stages both in patients and preclinical models, how microglia cells respond to altered neurodevelopmental dynamics remains not completely understood. To gain an insight on this issue, we have characterized patterns of microglia activation in the early postnatal cerebellum and young adult olfactory bulb of the hypomorphic Npc1nmf164 mouse model. Previous evidence has shown that both these areas display a number of anomalies affecting neuron and glial cell proliferation and differentiation, which largely anticipate cellular changes and clinical signs, raising our interest on how microglia interplay to these changes. Even so, to separate the contribution of cues provided by the dysfunctional microenvironment we have also studied microglia isolated from mice of increasing ages and cultured in vitro for 1 week. Our findings show that microglia of both cerebellum and olfactory bulb of Npc1nmf164 mice adopt an activated phenotype, characterized by increased cell proliferation, enlarged soma size and de-ramified processes, as well as a robust phagocytic activity, in a time- and space-specific manner. Enhanced phagocytosis associates with a profound remodeling of gene expression signatures towards gene products involved in chemotaxis, cell recognition and engulfment, including Cd68 and Trem2. These early changes in microglia morphology and activities are induced by region-specific developmental anomalies that likely anticipate alterations in neuronal connectivity. As a proof of concept, we show that microglia activation within the granule cell layer and glomerular layer of the olfactory bulb of Npc1nmf164 mice is associated with shortfalls in fine odor discrimination.
Collapse
Affiliation(s)
- Alessandro Rava
- Division of Neuroscience, Department of Psychology University La Sapienza Rome Italy
- PhD program in Behavioral Neuroscience University La Sapienza Rome Italy
| | - Piergiorgio La Rosa
- Division of Neuroscience, Department of Psychology University La Sapienza Rome Italy
- European Center for Brain Research IRCCS Fondazione Santa Lucia Rome Italy
| | - Giampiero Palladino
- Division of Neuroscience, Department of Psychology University La Sapienza Rome Italy
- PhD program in Behavioral Neuroscience University La Sapienza Rome Italy
| | - Jessica Dragotto
- Division of Neuroscience, Department of Psychology University La Sapienza Rome Italy
- PhD program in Behavioral Neuroscience University La Sapienza Rome Italy
| | - Antonio Totaro
- European Center for Brain Research IRCCS Fondazione Santa Lucia Rome Italy
| | - Jessica Tiberi
- Division of Neuroscience, Department of Psychology University La Sapienza Rome Italy
- PhD program in Behavioral Neuroscience University La Sapienza Rome Italy
| | - Sonia Canterini
- Division of Neuroscience, Department of Psychology University La Sapienza Rome Italy
| | - Sergio Oddi
- European Center for Brain Research IRCCS Fondazione Santa Lucia Rome Italy
- Faculty of Veterinary Medicine University of Teramo Teramo Italy
| | - Maria Teresa Fiorenza
- Division of Neuroscience, Department of Psychology University La Sapienza Rome Italy
- European Center for Brain Research IRCCS Fondazione Santa Lucia Rome Italy
| |
Collapse
|
7
|
Sharma S, Jeyaraman M, Muthu S. Role of stem cell therapy in neurosciences. ESSENTIALS OF EVIDENCE-BASED PRACTICE OF NEUROANESTHESIA AND NEUROCRITICAL CARE 2022:163-179. [DOI: 10.1016/b978-0-12-821776-4.00012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
8
|
Qiu X, Chen H, Feng D, Dong W. [G-protein coupled receptor Smo positively regulates proliferation and migration of adult neural stem cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1588-1592. [PMID: 34755677 DOI: 10.12122/j.issn.1673-4254.2021.10.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the role of G-protein coupled receptor Smoothened (Smo) in regulating proliferation and migration of adult neural stem cells (ANSCs) and explore the underlying mechanism. METHODS Cultured ANSCs were treated with purmorphamine (PM, an agonist of Smo) or cyclopamine (CPM, an inhibitor of Smo), and the changes in cell proliferation migration abilities were assessed using cell counting kit-8 (CCK8) assay and wound healing assay, respectively. The mRNA expressions of membrane receptor Patched 1 (Ptch1), Smo, glioma-associated oncogene homolog 1 (Gli1), axon guidance cue slit1 (Slit1) and brain-derived neurotrophic factor (BDNF) in the treated cells were detected using real-time quantitative PCR (RT-PCR). RESULTS PM significantly promoted the proliferation (P < 0.01) and migration of ANSCs (P < 0.01), and up-regulated the mRNA expressions of Ptch1, Smo, Gli1, Slit1 and BDNF. Treatment with CPM significantly inhibited the proliferation and migration of ANSCs. CONCLUSION Modulating Smo activity can positively regulate the proliferation and migration of ANSCs possibly by regulating the expressions of BDNF and Slit1.
Collapse
Affiliation(s)
- X Qiu
- Experiment Teaching and Administration Center, Southern Medical University, Guangzhou 510515, China
| | - H Chen
- Department of Neurosurgery, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - D Feng
- Institute of Oncology, Southern Medical University, Guangzhou 510515, China
| | - W Dong
- Experiment Teaching and Administration Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
9
|
Alonso A, Trujillo CM, Puelles L. Quail-chick grafting experiments corroborate that Tbr1-positive eminential prethalamic neurons migrate along three streams into hypothalamus, subpallium and septocommissural areas. Brain Struct Funct 2021; 226:759-785. [PMID: 33544184 PMCID: PMC7981335 DOI: 10.1007/s00429-020-02206-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
The prethalamic eminence (PThE), a diencephalic caudal neighbor of the telencephalon and alar hypothalamus, is frequently described in mammals and birds as a transient embryonic structure, undetectable in the adult brain. Based on descriptive developmental analysis of Tbr1 gene brain expression in chick embryos, we previously reported that three migratory cellular streams exit the PThE rostralward, targeting multiple sites in the hypothalamus, subpallium and septocommissural area, where eminential cells form distinct nuclei or disperse populations. These conclusions needed experimental corroboration. In this work, we used the homotopic quail-chick chimeric grafting procedure at stages HH10/HH11 to demonstrate by fate-mapping the three predicted tangential migration streams. Some chimeric brains were processed for Tbr1 in situ hybridization, for correlation with our previous approach. Evidence supporting all three postulated migration streams is presented. The results suggested a slight heterochrony among the juxtapeduncular (first), the peripeduncular (next), and the eminentio-septal (last) streams, each of which followed differential routes. A possible effect of such heterochrony on the differential selection of medial to lateral habenular hodologic targets by the migrated neurons is discussed.
Collapse
Affiliation(s)
- Antonia Alonso
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, School of Medicine, University of Murcia, 30100, Murcia, Spain. .,Biomedical Research Laboratory (LAIB), Health Campus, Murcia Biomedical Research Institute (IMIB-Arrixaca), El Palmar, 30120, Murcia, Spain.
| | - Carmen María Trujillo
- Department of Biochemistry, Microbiology, Cell Biology and Genetics, Faculty of Sciences, School of Biology, University of La Laguna, 38200, La Laguna, Canary Islands, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, School of Medicine, University of Murcia, 30100, Murcia, Spain.,Biomedical Research Laboratory (LAIB), Health Campus, Murcia Biomedical Research Institute (IMIB-Arrixaca), El Palmar, 30120, Murcia, Spain
| |
Collapse
|
10
|
Tirou L, Russo M, Faure H, Pellegrino G, Demongin C, Daynac M, Sharif A, Amosse J, Le Lay S, Denis R, Luquet S, Taouis M, Benomar Y, Ruat M. Sonic Hedgehog receptor Patched deficiency in astrocytes enhances glucose metabolism in mice. Mol Metab 2021; 47:101172. [PMID: 33513436 PMCID: PMC7893488 DOI: 10.1016/j.molmet.2021.101172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 01/06/2023] Open
Abstract
Objective Astrocytes are glial cells proposed as the main Sonic hedgehog (Shh)-responsive cells in the adult brain. Their roles in mediating Shh functions are still poorly understood. In the hypothalamus, astrocytes support neuronal circuits implicated in the regulation of energy metabolism. In this study, we investigated the impact of genetic activation of Shh signaling on hypothalamic astrocytes and characterized its effects on energy metabolism. Methods We analyzed the distribution of gene transcripts of the Shh pathway (Ptc, Gli1, Gli2, and Gli3) in astrocytes using single molecule fluorescence in situ hybridization combined with immunohistofluorescence of Shh peptides by Western blotting in the adult mouse hypothalamus. Based on the metabolic phenotype, we characterized Glast-CreERT2-YFP-Ptc−/− (YFP-Ptc−/−) mice and their controls over time and under a high-fat diet (HFD) to investigate the potential effects of conditional astrocytic deletion of the Shh receptor Patched (Ptc) on metabolic efficiency, insulin sensitivity, and systemic glucose metabolism. Molecular and biochemical assays were used to analyze the alteration of key pathways modulating energy metabolism, insulin sensitivity, glucose uptake, and inflammation. Primary astrocyte cultures were used to evaluate a potential role of Shh signaling in astrocytic glucose uptake. Results Shh peptides were the highest in the hypothalamic extracts of adult mice and a large population of hypothalamic astrocytes expressed Ptc and Gli1-3 mRNAs. Characterization of Shh signaling after conditional Ptc deletion in the YFP-Ptc−/− mice revealed heterogeneity in hypothalamic astrocyte populations. Interestingly, activation of Shh signaling in Glast+ astrocytes enhanced insulin responsiveness as evidenced by glucose and insulin tolerance tests. This effect was maintained over time and associated with lower blood insulin levels and also observed under a HFD. The YFP-Ptc−/− mice exhibited a lean phenotype with the absence of body weight gain and a marked reduction of white and brown adipose tissues accompanied by increased whole-body fatty acid oxidation. In contrast, food intake, locomotor activity, and body temperature were not altered. At the cellular level, Ptc deletion did not affect glucose uptake in primary astrocyte cultures. In the hypothalamus, activation of the astrocytic Shh pathway was associated with the upregulation of transcripts coding for the insulin receptor and liver kinase B1 (LKB1) after 4 weeks and the glucose transporter GLUT-4 after 32 weeks. Conclusions Here, we define hypothalamic Shh action on astrocytes as a novel master regulator of energy metabolism. In the hypothalamus, astrocytic Shh signaling could be critically involved in preventing both aging- and obesity-related metabolic disorders. Astrocytes exhibit differences in regulating the hedgehog signaling pathway. Deletion of Ptc in Glast+ cells prevents body weight gain and insulin resistance. Deletion of Ptc in Glast+ cells increases β oxidation. Central hedgehog signaling participates in the regulation of whole-body metabolism.
Collapse
Affiliation(s)
- Linda Tirou
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Mariagiovanna Russo
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Helene Faure
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Giuliana Pellegrino
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Clement Demongin
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Mathieu Daynac
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog (JPARC) - Lille Neurosciences & Cognition, F-59000, Lille, France
| | - Jeremy Amosse
- Univ. Angers SFR ICAT, F-49100, Angers, France; IRSET Laboratory, Inserm, UMR, 1085, Rennes, France
| | - Soazig Le Lay
- Univ. Angers SFR ICAT, F-49100, Angers, France; Univ. Nantes, CNRS, Inserm, Thorax Institut, F-44000, Nantes, France
| | - Raphaël Denis
- Univ. Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Serge Luquet
- Univ. Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Mohammed Taouis
- CNRS, Paris-Saclay University, UMR 9197, Neuroscience Paris-Saclay Institute, Molecular Neuroendocrinology of Food Intake, Orsay, France
| | - Yacir Benomar
- CNRS, Paris-Saclay University, UMR 9197, Neuroscience Paris-Saclay Institute, Molecular Neuroendocrinology of Food Intake, Orsay, France
| | - Martial Ruat
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France.
| |
Collapse
|
11
|
Kim Y, Lee J, Seppala M, Cobourne MT, Kim SH. Ptch2/Gas1 and Ptch1/Boc differentially regulate Hedgehog signalling in murine primordial germ cell migration. Nat Commun 2020; 11:1994. [PMID: 32332736 PMCID: PMC7181751 DOI: 10.1038/s41467-020-15897-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 04/01/2020] [Indexed: 12/24/2022] Open
Abstract
Gas1 and Boc/Cdon act as co-receptors in the vertebrate Hedgehog signalling pathway, but the nature of their interaction with the primary Ptch1/2 receptors remains unclear. Here we demonstrate, using primordial germ cell migration in mouse as a developmental model, that specific hetero-complexes of Ptch2/Gas1 and Ptch1/Boc mediate the process of Smo de-repression with different kinetics, through distinct modes of Hedgehog ligand reception. Moreover, Ptch2-mediated Hedgehog signalling induces the phosphorylation of Creb and Src proteins in parallel to Gli induction, identifying a previously unknown Ptch2-specific signal pathway. We propose that although Ptch1 and Ptch2 functionally overlap in the sequestration of Smo, the spatiotemporal expression of Boc and Gas1 may determine the outcome of Hedgehog signalling through compartmentalisation and modulation of Smo-downstream signalling. Our study identifies the existence of a divergent Hedgehog signal pathway mediated by Ptch2 and provides a mechanism for differential interpretation of Hedgehog signalling in the germ cell niche. How co-receptors Gas1 and Boc interact with Ptch1/2 receptors and regulate Hh signalling is unclear. Here, the authors demonstrate that the spatiotemporal expression of Gas1 and Boc determines how Hh signalling affects the dynamic migration of murine primordial germ cells.
Collapse
Affiliation(s)
- Yeonjoo Kim
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Jiyoung Lee
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Maisa Seppala
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences King's College London Floor 27, Guy's Hospital, London, SE1 9RT, UK
| | - Martyn T Cobourne
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences King's College London Floor 27, Guy's Hospital, London, SE1 9RT, UK
| | - Soo-Hyun Kim
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.
| |
Collapse
|
12
|
Tirou L, Russo M, Faure H, Pellegrino G, Sharif A, Ruat M. C9C5 positive mature oligodendrocytes are a source of Sonic Hedgehog in the mouse brain. PLoS One 2020; 15:e0229362. [PMID: 32078657 PMCID: PMC7032736 DOI: 10.1371/journal.pone.0229362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
In the mature rodent brain, Sonic Hedgehog (Shh) signaling regulates stem and progenitor cell maintenance, neuronal and glial circuitry and brain repair. However, the sources and distribution of Shh mediating these effects are still poorly characterized. Here, we report in the adult mouse brain, a broad expression pattern of Shh recognized by the specific monoclonal C9C5 antibody in a subset (11–12%) of CC1+ mature oligodendrocytes that do not express carbonic anhydrase II. These cells express also Olig2 and Sox10, two oligodendrocyte lineage-specific markers, but not PDGFRα, a marker of oligodendrocyte progenitors. In agreement with oligodendroglial cells being a source of Shh in the adult mouse brain, we identify Shh transcripts by single molecule fluorescent in situ hybridization in a subset of cells expressing Olig2 and Sox10 mRNAs. These findings also reveal that Shh expression is more extensive than originally reported. The Shh-C9C5-associated signal labels the oligodendroglial cell body and decorates by intense puncta the processes. C9C5+ cells are distributed in a grid-like manner. They constitute small units that could deliver locally Shh to its receptor Patched expressed in GFAP+ and S100β+ astrocytes, and in HuC/D+ neurons as shown in PtcLacZ/+ reporter mice. Postnatally, C9C5 immunoreactivity overlaps the myelination peak that occurs between P10 and P20 and is down regulated during ageing. Thus, our data suggest that C9C5+CC1+ oligodendroglial cells are a source of Shh in the mouse postnatal brain.
Collapse
Affiliation(s)
- Linda Tirou
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | - Mariagiovanna Russo
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | - Helene Faure
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | - Giuliana Pellegrino
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | | | - Martial Ruat
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
- * E-mail:
| |
Collapse
|
13
|
Dynamic Changes in Ultrastructure of the Primary Cilium in Migrating Neuroblasts in the Postnatal Brain. J Neurosci 2019; 39:9967-9988. [PMID: 31685650 DOI: 10.1523/jneurosci.1503-19.2019] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 10/08/2019] [Accepted: 10/24/2019] [Indexed: 11/21/2022] Open
Abstract
New neurons, referred to as neuroblasts, are continuously generated in the ventricular-subventricular zone of the brain throughout an animal's life. These neuroblasts are characterized by their unique potential for proliferation, formation of chain-like cell aggregates, and long-distance and high-speed migration through the rostral migratory stream (RMS) toward the olfactory bulb (OB), where they decelerate and differentiate into mature interneurons. The dynamic changes of ultrastructural features in postnatal-born neuroblasts during migration are not yet fully understood. Here we report the presence of a primary cilium, and its ultrastructural morphology and spatiotemporal dynamics, in migrating neuroblasts in the postnatal RMS and OB. The primary cilium was observed in migrating neuroblasts in the postnatal RMS and OB in male and female mice and zebrafish, and a male rhesus monkey. Inhibition of intraflagellar transport molecules in migrating neuroblasts impaired their ciliogenesis and rostral migration toward the OB. Serial section transmission electron microscopy revealed that each migrating neuroblast possesses either a pair of centrioles or a basal body with an immature or mature primary cilium. Using immunohistochemistry, live imaging, and serial block-face scanning electron microscopy, we demonstrate that the localization and orientation of the primary cilium are altered depending on the mitotic state, saltatory migration, and deceleration of neuroblasts. Together, our results highlight a close mutual relationship between spatiotemporal regulation of the primary cilium and efficient chain migration of neuroblasts in the postnatal brain.SIGNIFICANCE STATEMENT Immature neurons (neuroblasts) generated in the postnatal brain have a mitotic potential and migrate in chain-like cell aggregates toward the olfactory bulb. Here we report that migrating neuroblasts possess a tiny cellular protrusion called a primary cilium. Immunohistochemical studies with zebrafish, mouse, and monkey brains suggest that the presence of the primary cilium in migrating neuroblasts is evolutionarily conserved. Ciliogenesis in migrating neuroblasts in the rostral migratory stream is suppressed during mitosis and promoted after cell cycle exit. Moreover, live imaging and 3D electron microscopy revealed that ciliary localization and orientation change during saltatory movement of neuroblasts. Our results reveal highly organized dynamics in maturation and positioning of the primary cilium during neuroblast migration that underlie saltatory movement of postnatal-born neuroblasts.
Collapse
|
14
|
Adult Neurogenesis in the Subventricular Zone and Its Regulation After Ischemic Stroke: Implications for Therapeutic Approaches. Transl Stroke Res 2019; 11:60-79. [DOI: 10.1007/s12975-019-00717-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
|
15
|
Del Giovane A, Ragnini-Wilson A. Targeting Smoothened as a New Frontier in the Functional Recovery of Central Nervous System Demyelinating Pathologies. Int J Mol Sci 2018; 19:E3677. [PMID: 30463396 PMCID: PMC6274747 DOI: 10.3390/ijms19113677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
Abstract
Myelin sheaths on vertebrate axons provide protection, vital support and increase the speed of neuronal signals. Myelin degeneration can be caused by viral, autoimmune or genetic diseases. Remyelination is a natural process that restores the myelin sheath and, consequently, neuronal function after a demyelination event, preventing neurodegeneration and thereby neuron functional loss. Pharmacological approaches to remyelination represent a promising new frontier in the therapy of human demyelination pathologies and might provide novel tools to improve adaptive myelination in aged individuals. Recent phenotypical screens have identified agonists of the atypical G protein-coupled receptor Smoothened and inhibitors of the glioma-associated oncogene 1 as being amongst the most potent stimulators of oligodendrocyte precursor cell (OPC) differentiation in vitro and remyelination in the central nervous system (CNS) of mice. Here, we discuss the current state-of-the-art of studies on the role of Sonic Hedgehog reactivation during remyelination, referring readers to other reviews for the role of Hedgehog signaling in cancer and stem cell maintenance.
Collapse
Affiliation(s)
- Alice Del Giovane
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| | - Antonella Ragnini-Wilson
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| |
Collapse
|
16
|
Casillas C, Roelink H. Gain-of-function Shh mutants activate Smo cell-autonomously independent of Ptch1/2 function. Mech Dev 2018; 153:30-41. [PMID: 30144507 PMCID: PMC6165682 DOI: 10.1016/j.mod.2018.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/25/2022]
Abstract
Sonic Hedgehog (Shh) signaling is characterized by non-cell autonomy; cells expressing Shh do not respond to the ligand. Here, we identify several Shh mutations that can activate the Hedgehog (Hh) pathway cell-autonomously. Cell-autonomous pathway activation requires the extracellular cysteine rich domain of Smoothened, but is otherwise independent of the Shh receptors Patched1 and -2. Many of the Shh mutants that gain activity fail to undergo auto processing resulting in the perdurance of the Shh pro-peptide, a form of Shh that is sufficient to activate the Hh response cell-autonomously. Our results demonstrate that Shh is capable of activating the Hh pathway via Smoothened, independently of Patched1/2, and that it harbors an intrinsic mechanism that prevents cell-autonomous activation of the Shh response.
Collapse
Affiliation(s)
- Catalina Casillas
- Department of Molecular and Cell Biology, 16 Barker Hall, 3204, University of California, Berkeley, CA 94720, USA
| | - Henk Roelink
- Department of Molecular and Cell Biology, 16 Barker Hall, 3204, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
17
|
Adams KV, Morshead CM. Neural stem cell heterogeneity in the mammalian forebrain. Prog Neurobiol 2018; 170:2-36. [PMID: 29902499 DOI: 10.1016/j.pneurobio.2018.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 05/23/2018] [Accepted: 06/07/2018] [Indexed: 12/21/2022]
Abstract
The brain was long considered an organ that underwent very little change after development. It is now well established that the mammalian central nervous system contains neural stem cells that generate progeny that are capable of making new neurons, astrocytes, and oligodendrocytes throughout life. The field has advanced rapidly as it strives to understand the basic biology of these precursor cells, and explore their potential to promote brain repair. The purpose of this review is to present current knowledge about the diversity of neural stem cells in vitro and in vivo, and highlight distinctions between neural stem cell populations, throughout development, and within the niche. A comprehensive understanding of neural stem cell heterogeneity will provide insights into the cellular and molecular regulation of neural development and lifelong neurogenesis, and will guide the development of novel strategies to promote regeneration and neural repair.
Collapse
Affiliation(s)
- Kelsey V Adams
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada.
| | - Cindi M Morshead
- Institute of Medical Science, Terrence Donnelly Centre, University of Toronto, Toronto ON, M5S 3E2, Canada; Department of Surgery, Division of Anatomy, Canada; Institute of Biomaterials and Biomedical Engineering, Canada; Rehabilitation Science Institute, University of Toronto, Canada.
| |
Collapse
|
18
|
Liu X, Wang Y, Liu F, Zhang M, Song H, Zhou B, Lo CW, Tong S, Hu Z, Zhang Z. Wdpcp promotes epicardial EMT and epicardium-derived cell migration to facilitate coronary artery remodeling. Sci Signal 2018; 11:11/519/eaah5770. [PMID: 29487191 DOI: 10.1126/scisignal.aah5770] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
During coronary vasculature development, endothelial cells enclose the embryonic heart to form the primitive coronary plexus. This structure is remodeled upon recruitment of epicardial cells that may undergo epithelial-mesenchymal transition (EMT) to enable migration and that give rise to smooth muscle cells. In mice expressing a loss-of-function mutant form of Wdpcp, a gene involved in ciliogenesis, the enclosure of the surface of the heart by the subepicardial coronary plexus was accelerated because of enhanced chemotactic responses to Shh. Coronary arteries, but not coronary veins in Wdpcp mutant mice, showed reduced smooth muscle cell coverage. In addition, Wdpcp mutant hearts had reduced expression of EMT and mesenchymal markers and had fewer epicardium-derived cells (EPDCs) that showed impaired migration. Epicardium-specific deletion of Wdpcp recapitulated the coronary artery defect of the Wdpcp mutant. Thus, Wdpcp promotes epithelial EMT and EPDC migration, processes that are required for remodeling of the coronary primitive plexus. The Wdpcp mutant mice will be a useful tool to dissect the molecular mechanisms that govern the remodeling of the primitive plexus during coronary development.
Collapse
Affiliation(s)
- Xiangyang Liu
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ye Wang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Feng Liu
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Min Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hejie Song
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Bin Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15201, USA
| | - Shilu Tong
- Department of Clinical Epidemiology and Biostatistics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhenlei Hu
- Department of Cardiovascular Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zhen Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
19
|
Cerebellar granule cell replenishment postinjury by adaptive reprogramming of Nestin + progenitors. Nat Neurosci 2017; 20:1361-1370. [PMID: 28805814 PMCID: PMC5614835 DOI: 10.1038/nn.4621] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 07/11/2017] [Indexed: 12/16/2022]
Abstract
Regeneration of several organs involves adaptive reprogramming of progenitors, however, the intrinsic capacity of the developing brain to replenish lost cells remains largely unknown. In this study, we discovered that the developing cerebellum has unappreciated progenitor plasticity, since it undergoes near full growth and functional recovery following acute depletion of granule cells, the most plentiful neuron population in the brain. We demonstrate that following postnatal ablation of granule cell progenitors, Nestin-expressing progenitors (NEPs) specified during mid-embryogenesis to produce astroglia and interneurons, switch their fate and generate granule neurons in mice. Moreover, Hedgehog-signaling in two NEP populations is crucial not only for the compensatory replenishment of granule neurons but also to scale interneuron and astrocyte numbers. Thus we provide insights into the mechanisms underlying robustness of circuit formation in the cerebellum, and speculate that adaptive reprogramming of progenitors in other brain regions plays a greater role than appreciated in developmental regeneration.
Collapse
|
20
|
Faissner A, Roll L, Theocharidis U. Tenascin-C in the matrisome of neural stem and progenitor cells. Mol Cell Neurosci 2017; 81:22-31. [DOI: 10.1016/j.mcn.2016.11.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 01/16/2023] Open
|
21
|
Jin Y, Barnett A, Zhang Y, Yu X, Luo Y. Poststroke Sonic Hedgehog Agonist Treatment Improves Functional Recovery by Enhancing Neurogenesis and Angiogenesis. Stroke 2017; 48:1636-1645. [PMID: 28487338 DOI: 10.1161/strokeaha.117.016650] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/20/2017] [Accepted: 03/30/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE Because of the limitation in treatment window of the r-tPA (recombinant tissue-type plasminogen activator), the development of delayed treatment for stroke is needed. In this study, we examined the efficacy of delayed poststroke treatment (post 3-8 days) of the sonic hedgehog pathway agonist on functional recovery and the underlying mechanisms. METHODS We evaluated functional recovery at 1 month after stroke using locomotion analysis and Barnes maze test for cognitive function. We used a genetically inducible neural stem cell-specific reporter mouse line (nestin-CreERT2-R26R-YFP) to label and track their proliferation, survival, and differentiation in ischemic brain. Brain tissue damage, angiogenesis, and cerebral blood flow recovery was evaluated using magnetic resonance imaging techniques and immunostaining. RESULTS Our results show that delayed treatment of sonic hedgehog pathway agonist in stroke mice results in enhanced functional recovery both in locomotor function and in cognitive function at 1 month after stroke. Furthermore, using the Nestincre-ERT2-YFP mice, we showed that poststroke sonic hedgehog pathway agonist treatment increased surviving newly born cells derived from both subventricular zone and subgranular zone neural stem cells, total surviving DCX+ (Doublecortin) neuroblast cells, and neurons (NeuN+/YFP+) in the ischemic brain. Sonic hedgehog pathway agonist treatment also improved the brain tissue repair in ischemic region supported by our T2-weighted magnetic resonance imaging, cerebral blood flow map by arterial spin labeling, and immunohistochemistry (α-smooth muscle actin and CD31 immunostaining). CONCLUSIONS These data confirm an important role for the hedgehog pathway in poststroke brain repair and functional recovery, suggesting a prolonged treatment window for potential treatment strategy to modulate sonic hedgehog pathway after stroke.
Collapse
Affiliation(s)
- Yongming Jin
- From the Department of Neurological Surgery (Y.J., A.B., Y.L.) and Department of Biomedical Engineering (Y.Z., X.Y.), Case Western Reserve University, Cleveland, OH
| | - Austin Barnett
- From the Department of Neurological Surgery (Y.J., A.B., Y.L.) and Department of Biomedical Engineering (Y.Z., X.Y.), Case Western Reserve University, Cleveland, OH
| | - Yifan Zhang
- From the Department of Neurological Surgery (Y.J., A.B., Y.L.) and Department of Biomedical Engineering (Y.Z., X.Y.), Case Western Reserve University, Cleveland, OH
| | - Xin Yu
- From the Department of Neurological Surgery (Y.J., A.B., Y.L.) and Department of Biomedical Engineering (Y.Z., X.Y.), Case Western Reserve University, Cleveland, OH
| | - Yu Luo
- From the Department of Neurological Surgery (Y.J., A.B., Y.L.) and Department of Biomedical Engineering (Y.Z., X.Y.), Case Western Reserve University, Cleveland, OH.
| |
Collapse
|
22
|
Abstract
Olfactory axons project from nasal epithelium to the primitive telencephalon before olfactory bulbs form. Olfactory bulb neurons do not differentiate in situ but arrive via the rostral migratory stream. Synaptic glomeruli and concentric laminar architecture are unlike other cortices. Fetal olfactory maturation of neuronal differentiation, synaptogenesis, and myelination remains incomplete at term and have a protracted course of postnatal development. The olfactory ventricular recess involutes postnatally but dilates in congenital hydrocephalus. Olfactory bulb, tract and epithelium are repositories of progenitor stem cells in fetal and adult life. Diverse malformations of the olfactory bulb can be diagnosed by clinical examination, imaging, and neuropathologically. Cellular markers of neuronal differentiation and synaptogenesis demonstrate immaturity of the olfactory system at birth, previously believed by histology alone to occur early in fetal life. Immaturity does not preclude function.
Collapse
Affiliation(s)
- Harvey B Sarnat
- 1 Department of Paediatrics, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.,2 Department of Pathology and Laboratory Medicine (Neuropathology), University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.,3 Department of Clinical Neurosciences, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Laura Flores-Sarnat
- 1 Department of Paediatrics, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada.,3 Department of Clinical Neurosciences, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Kaneko N, Sawada M, Sawamoto K. Mechanisms of neuronal migration in the adult brain. J Neurochem 2017; 141:835-847. [DOI: 10.1111/jnc.14002] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/06/2017] [Accepted: 02/21/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Naoko Kaneko
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medial Sciences; Nagoya Aichi Japan
| | - Masato Sawada
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medial Sciences; Nagoya Aichi Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology; Nagoya City University Graduate School of Medial Sciences; Nagoya Aichi Japan
- Division of Neural Development and Regeneration; National Institute for Physiological Sciences; Okazaki Aichi Japan
| |
Collapse
|
24
|
Improved smell function with increased nasal mucus sonic hedgehog in hyposmic patients after treatment with oral theophylline. Am J Otolaryngol 2017; 38:143-147. [PMID: 27923495 DOI: 10.1016/j.amjoto.2016.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/22/2016] [Indexed: 01/01/2023]
Abstract
PURPOSE We previously demonstrated the presence of sonic hedgehog (Shh) in nasal mucus in normal subjects and in patients with smell loss (hyposmia). Nasal mucus Shh levels were found significantly diminished in untreated hyposmic patients of multiple etiologies. Since treatment with oral theophylline has been previously associated with improvement in smell function we wished to study if such treatment increased nasal mucus Shh as well as improved smell function in patients with hyposmia. METHODS Forty-four patients with hyposmia of several etiologies were evaluated for changes in hyposmia by subjective measurements of smell, taste and flavor perception and by olfactometry. Measurements of nasal mucus Shh were made in relationship to each set of sensory measurements. Patients were treated with oral theophylline at doses of 200-800mg for periods of 2-10months with sensory function, nasal mucus Shh and serum theophylline levels evaluated at these time intervals. Nasal mucus Shh measurements were made with a sensitive spectrophotometric ELISA assay and theophylline with a fluorometric assay. RESULTS There was consistent, significant improvement in subjective responses in smell, taste and flavor perception and in olfactometry associated with increased nasal mucus Shh and serum theophylline after theophylline treatment. CONCLUSIONS Improvement in smell function and in nasal mucus Shh was positively correlated in a dose-response relationship after treatment with oral theophylline. Results are consistent with a successful role for theophylline in improvement of smell function in hyposmic patients of multiple etiologies associated with increased nasal mucus Shh which can act as a biochemical marker for smell function.
Collapse
|
25
|
Liu C, Bousman CA, Pantelis C, Skafidas E, Zhang D, Yue W, Everall IP. Pathway-wide association study identifies five shared pathways associated with schizophrenia in three ancestral distinct populations. Transl Psychiatry 2017; 7:e1037. [PMID: 28221366 PMCID: PMC5438037 DOI: 10.1038/tp.2017.8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/30/2016] [Indexed: 02/08/2023] Open
Abstract
Genome-wide association studies have confirmed the polygenic nature of schizophrenia and suggest that there are hundreds or thousands of alleles associated with increased liability for the disorder. However, the generalizability of any one allelic marker of liability is remarkably low and has bred the notion that schizophrenia may be better conceptualized as a pathway(s) disorder. Here, we empirically tested this notion by conducting a pathway-wide association study (PWAS) encompassing 255 experimentally validated Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways among 5033 individuals diagnosed with schizophrenia and 5332 unrelated healthy controls across three distinct ethnic populations; European-American (EA), African-American (AA) and Han Chinese (CH). We identified 103, 74 and 87 pathways associated with schizophrenia liability in the EA, CH and AA populations, respectively. About half of these pathways were uniquely associated with schizophrenia liability in each of the three populations. Five pathways (serotonergic synapse, ubiquitin mediated proteolysis, hedgehog signaling, adipocytokine signaling and renin secretion) were shared across all three populations and the single-nucleotide polymorphism sets representing these five pathways were enriched for single-nucleotide polymorphisms with regulatory function. Our findings provide empirical support for schizophrenia as a pathway disorder and suggest schizophrenia is not only a polygenic but likely also a poly-pathway disorder characterized by both genetic and pathway heterogeneity.
Collapse
Affiliation(s)
- C Liu
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
| | - C A Bousman
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of General Practice, The University of Melbourne, Parkville, VIC, Australia
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - C Pantelis
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Electrical and Electronic Engineering, Centre for Neural Engineering (CfNE), University of Melbourne, Carlton South, VIC, Australia
- NorthWestern Mental Health, Melbourne, VIC, Australia
| | - E Skafidas
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Electrical and Electronic Engineering, Centre for Neural Engineering (CfNE), University of Melbourne, Carlton South, VIC, Australia
| | - D Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
- Peking-Tsinghua Joint Center for Life Sciences/PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - W Yue
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, China
| | - I P Everall
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Electrical and Electronic Engineering, Centre for Neural Engineering (CfNE), University of Melbourne, Carlton South, VIC, Australia
- NorthWestern Mental Health, Melbourne, VIC, Australia
| |
Collapse
|
26
|
Chew LJ, DeBoy CA. Pharmacological approaches to intervention in hypomyelinating and demyelinating white matter pathology. Neuropharmacology 2016; 110:605-625. [PMID: 26116759 PMCID: PMC4690794 DOI: 10.1016/j.neuropharm.2015.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 06/10/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022]
Abstract
White matter disease afflicts both developing and mature central nervous systems. Both cell intrinsic and extrinsic dysregulation result in profound changes in cell survival, axonal metabolism and functional performance. Experimental models of developmental white matter (WM) injury and demyelination have not only delineated mechanisms of signaling and inflammation, but have also paved the way for the discovery of pharmacological approaches to intervention. These reagents have been shown to enhance protection of the mature oligodendrocyte cell, accelerate progenitor cell recruitment and/or differentiation, or attenuate pathological stimuli arising from the inflammatory response to injury. Here we highlight reports of studies in the CNS in which compounds, namely peptides, hormones, and small molecule agonists/antagonists, have been used in experimental animal models of demyelination and neonatal brain injury that affect aspects of excitotoxicity, oligodendrocyte development and survival, and progenitor cell function, and which have been demonstrated to attenuate damage and improve WM protection in experimental models of injury. The molecular targets of these agents include growth factor and neurotransmitter receptors, morphogens and their signaling components, nuclear receptors, as well as the processes of iron transport and actin binding. By surveying the current evidence in non-immune targets of both the immature and mature WM, we aim to better understand pharmacological approaches modulating endogenous oligodendroglia that show potential for success in the contexts of developmental and adult WM pathology. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| | - Cynthia A DeBoy
- Biology Department, Trinity Washington University, Washington, DC, USA
| |
Collapse
|
27
|
Daynac M, Tirou L, Faure H, Mouthon MA, Gauthier LR, Hahn H, Boussin FD, Ruat M. Hedgehog Controls Quiescence and Activation of Neural Stem Cells in the Adult Ventricular-Subventricular Zone. Stem Cell Reports 2016; 7:735-748. [PMID: 27666792 PMCID: PMC5063572 DOI: 10.1016/j.stemcr.2016.08.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/23/2016] [Accepted: 08/23/2016] [Indexed: 01/20/2023] Open
Abstract
Identifying the mechanisms controlling quiescence and activation of neural stem cells (NSCs) is crucial for understanding brain repair. Here, we demonstrate that Hedgehog (Hh) signaling actively regulates different pools of quiescent and proliferative NSCs in the adult ventricular-subventricular zone (V-SVZ), one of the main brain neurogenic niches. Specific deletion of the Hh receptor Patched in NSCs during adulthood upregulated Hh signaling in quiescent NSCs, progressively leading to a large accumulation of these cells in the V-SVZ. The pool of non-neurogenic astrocytes was not modified, whereas the activated NSC pool increased after a short period, before progressively becoming exhausted. We also showed that Sonic Hedgehog regulates proliferation of activated NSCs in vivo and shortens both their G1 and S-G2/M phases in culture. These data demonstrate that Hh orchestrates the balance between quiescent and activated NSCs, with important implications for understanding adult neurogenesis under normal homeostatic conditions or during injury.
Collapse
Affiliation(s)
- Mathieu Daynac
- CNRS, UMR-9197, Neuroscience Paris-Saclay Institute, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Linda Tirou
- CNRS, UMR-9197, Neuroscience Paris-Saclay Institute, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Hélène Faure
- CNRS, UMR-9197, Neuroscience Paris-Saclay Institute, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Marc-André Mouthon
- CEA DSV iRCM SCSR, Laboratoire de Radiopathologie, 92265 Fontenay-aux-Roses, France; INSERM, UMR 967, 92265 Fontenay-aux-Roses, France; Université Paris Sud, UMR 967, 92265 Fontenay-aux-Roses, France; Université Paris Diderot, Sorbonne Paris Cité, UMR 967, 92265 Fontenay-aux-Roses, France
| | - Laurent R Gauthier
- CEA DSV iRCM SCSR, Laboratoire de Radiopathologie, 92265 Fontenay-aux-Roses, France; INSERM, UMR 967, 92265 Fontenay-aux-Roses, France; Université Paris Sud, UMR 967, 92265 Fontenay-aux-Roses, France; Université Paris Diderot, Sorbonne Paris Cité, UMR 967, 92265 Fontenay-aux-Roses, France
| | - Heidi Hahn
- Tumor Genetics Group, Institute of Human Genetics, University Medical Center, 37073 Göttingen, Germany
| | - François D Boussin
- CEA DSV iRCM SCSR, Laboratoire de Radiopathologie, 92265 Fontenay-aux-Roses, France; INSERM, UMR 967, 92265 Fontenay-aux-Roses, France; Université Paris Sud, UMR 967, 92265 Fontenay-aux-Roses, France; Université Paris Diderot, Sorbonne Paris Cité, UMR 967, 92265 Fontenay-aux-Roses, France.
| | - Martial Ruat
- CNRS, UMR-9197, Neuroscience Paris-Saclay Institute, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France.
| |
Collapse
|
28
|
Sarnat HB, Yu W. Maturation and Dysgenesis of the Human Olfactory Bulb. Brain Pathol 2016; 26:301-18. [PMID: 26096058 PMCID: PMC8028954 DOI: 10.1111/bpa.12275] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/09/2015] [Indexed: 12/22/2022] Open
Abstract
The olfactory bulb with its unique architecture was studied for neuronal maturation in human fetuses. Neuroblasts stream into the olfactory bulb from the rostral telencephalon and secondarily migrate radially. The transitory olfactory ventricular recess regresses postnatally. Olfactory is the only sensory system without thalamic projections but incorporates intrinsic thalamic equivalents. The bulb is a repository of progenitor cells. Maturation of the bulb and tract was studied in 18 normal human fetuses of 16-41 weeks gestation; mid-gestational twins with hydrocephalus; 7 arrhinencephaly/holoprosencephaly; 2 olfactory dysgeneses. Multiple immunoreactivities were performed. Synaptophysin around mitral neurons, in a few synaptic glomeruli and concentric lamination of the outer granular layer, was seen at 16 weeks. Outer granular neurons exhibited NeuN at 16 weeks, only 2/3 were reactive at term. Concentric alternating sheets of granular neurons and their dendrodendritic synapses are seen during maturation. Calretinin reactivity is seen in neurons and neurites, primary olfactory nerve axons, periglomerular cells and neuroepithelial cells surrounding the ventricular recess; reactivity occurs later in synaptic glomeruli than with synaptophysin; not all glomeruli are strongly reactive even at term. Nestin- and vimentin-reactive bipolar progenitor cells were demonstrated at all ages and extend into the olfactory tract. Myelin is demonstrated by Luxol fast blue (LFB) only postnatally. In hydrocephalus, the olfactory recess is dilated. Mitral cell dispersion, disrupted glomeruli, heterotopia and maturational delay are seen in some dysgeneses. Malformations exhibit unique findings. Fusion of hypoplastic bulbs can occur. Abnormal architecture is seen in hemimegalencephaly. More documentation of olfactory dysgenesis is needed in other major brain malformations.
Collapse
Affiliation(s)
- Harvey B. Sarnat
- Department of PaediatricsUniversity of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
- Department of Pathology and Laboratory Medicine (Neuropathology)University of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
- Department of Clinical NeurosciencesUniversity of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
| | - Weiming Yu
- Department of PaediatricsUniversity of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
- Department of Pathology and Laboratory Medicine (Paediatric Pathology)University of Calgary Faculty of Medicine and Alberta Children's Hospital Research InstituteCalgaryABCanada
| |
Collapse
|
29
|
Tolosa EJ, Fernández-Zapico ME, Battiato NL, Rovasio RA. Sonic hedgehog is a chemotactic neural crest cell guide that is perturbed by ethanol exposure. Eur J Cell Biol 2016; 95:136-52. [DOI: 10.1016/j.ejcb.2016.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 01/23/2016] [Accepted: 02/17/2016] [Indexed: 12/12/2022] Open
|
30
|
Benetti EM, Gunnewiek MK, van Blitterswijk CA, Julius Vancso G, Moroni L. Mimicking natural cell environments: design, fabrication and application of bio-chemical gradients on polymeric biomaterial substrates. J Mater Chem B 2016; 4:4244-4257. [DOI: 10.1039/c6tb00947f] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gradients of biomolecules on synthetic, solid substrates can efficiently mimic the natural, graded variation of properties of the extracellular matrix (ECM).
Collapse
Affiliation(s)
- Edmondo M. Benetti
- Department of Materials Science and Technology of Polymers
- MESA+ Institute for Nanotechnology
- University of Twente
- 7500 AE Enschede
- The Netherlands
| | - Michel Klein Gunnewiek
- Department of Materials Science and Technology of Polymers
- MESA+ Institute for Nanotechnology
- University of Twente
- 7500 AE Enschede
- The Netherlands
| | - Clemens A. van Blitterswijk
- Department of Complex Tissue Regeneration
- MERLN Institute for Technology Inspired Regenerative Medicine
- Maastricht University
- 6200 MD Maastricht
- The Netherlands
| | - G. Julius Vancso
- Department of Materials Science and Technology of Polymers
- MESA+ Institute for Nanotechnology
- University of Twente
- 7500 AE Enschede
- The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration
- MERLN Institute for Technology Inspired Regenerative Medicine
- Maastricht University
- 6200 MD Maastricht
- The Netherlands
| |
Collapse
|
31
|
Porcu G, Serone E, De Nardis V, Di Giandomenico D, Lucisano G, Scardapane M, Poma A, Ragnini-Wilson A. Clobetasol and Halcinonide Act as Smoothened Agonists to Promote Myelin Gene Expression and RxRγ Receptor Activation. PLoS One 2015; 10:e0144550. [PMID: 26658258 PMCID: PMC4689554 DOI: 10.1371/journal.pone.0144550] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022] Open
Abstract
One of the causes of permanent disability in chronic multiple sclerosis patients is the inability of oligodendrocyte progenitor cells (OPCs) to terminate their maturation program at lesions. To identify key regulators of myelin gene expression acting at the last stages of OPC maturation we developed a drug repositioning strategy based on the mouse immortalized oligodendrocyte (OL) cell line Oli-neu brought to the premyelination stage by stably expressing a key factor regulating the last stages of OL maturation. The Prestwick Chemical Library® of 1,200 FDA-approved compound(s) was repositioned at three dosages based on the induction of Myelin Basic Protein (MBP) expression. Drug hits were further validated using dosage-dependent reproducibility tests and biochemical assays. The glucocorticoid class of compounds was the most highly represented and we found that they can be divided in three groups according to their efficacy on MBP up-regulation. Since target identification is crucial before bringing compounds to the clinic, we searched for common targets of the primary screen hits based on their known chemical-target interactomes, and the pathways predicted by top ranking compounds were validated using specific inhibitors. Two of the top ranking compounds, Halcinonide and Clobetasol, act as Smoothened (Smo) agonists to up-regulate myelin gene expression in the Oli-neuM cell line. Further, RxRγ activation is required for MBP expression upon Halcinonide and Clobetasol treatment. These data indicate Clobetasol and Halcinonide as potential promyelinating drugs and also provide a mechanistic understanding of their mode of action in the pathway leading to myelination in OPCs. Furthermore, our classification of glucocorticoids with respect to MBP expression provides important novel insights into their effects in the CNS and a rational criteria for their choice in combinatorial therapies in de-myelinating diseases.
Collapse
Affiliation(s)
- Giampiero Porcu
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
| | - Eliseo Serone
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L’Aquila, Italy
| | - Velia De Nardis
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
| | - Daniele Di Giandomenico
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
| | - Giuseppe Lucisano
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
- Dipartimento di Scienze Mediche di Base, Neuroscienze ed Organi di Senso, Università di Bari Aldo Moro, Bari, Italy
| | - Marco Scardapane
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
| | - Anna Poma
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L’Aquila, Italy
| | - Antonella Ragnini-Wilson
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Department of Translational Pharmacology, Fondazione Mario Negri Sud, S. Maria Imbaro (CH), Italy
- * E-mail:
| |
Collapse
|
32
|
Zhu M, Feng Y, Dangelmajer S, Guerrero-Cázares H, Chaichana KL, Smith CL, Levchenko A, Lei T, Quiñones-Hinojosa A. Human cerebrospinal fluid regulates proliferation and migration of stem cells through insulin-like growth factor-1. Stem Cells Dev 2015; 24:160-71. [PMID: 25265906 DOI: 10.1089/scd.2014.0076] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) and neural progenitor cells (NPCs) have been regarded for their clinical therapeutic potential for central nervous system (CNS) pathologies. Their potential utility is a result of their intrinsic ability to repair damaged tissues, deliver therapeutic proteins, and migrate to sites of pathology within the brain. However, it remains unclear whether the CNS promotes any changes in these potential therapeutic cells, which would be critical to understand before clinical application. A major component of the CNS is cerebrospinal fluid (CSF). Therefore, the aim of this study was to evaluate the influence that human CSF has on the function of human adipose-derived MSCs (hAMSCs) and human fetal-derived NPCs (hfNPCs) in regard to cell proliferation, survival, and migration. This study demonstrated that human noncancerous CSF promoted proliferation and inhibited apoptosis of hAMSCs and hfNPCs. Preculturing these stem cells in human CSF also increased their migratory speed and distance traveled. Furthermore, insulin-like growth factor-1 (IGF-1) in human CSF enhanced the migration capacity and increased the expression of C-X-C chemokine receptor type 4 (CXCR4) in both stem cell types. These current findings highlight a simple and natural way in which human CSF can enhance the proliferation, migration, and viability of human exogenous primary hAMSCs and hfNPCs. This study may provide insight into improving the clinical efficacy of stem cells for the treatment of CNS pathologies.
Collapse
Affiliation(s)
- Mingxin Zhu
- 1 Department of Neurosurgery and Oncology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Forrest CM, McNair K, Pisar M, Khalil OS, Darlington LG, Stone TW. Altered hippocampal plasticity by prenatal kynurenine administration, kynurenine-3-monoxygenase (KMO) deletion or galantamine. Neuroscience 2015; 310:91-105. [PMID: 26365611 PMCID: PMC4642643 DOI: 10.1016/j.neuroscience.2015.09.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/11/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022]
Abstract
Glutamate receptors sensitive to N-methyl-D-aspartate (NMDA) are involved in embryonic brain development but their activity may be modulated by the kynurenine pathway of tryptophan metabolism which includes an agonist (quinolinic acid) and an antagonist (kynurenic acid) at these receptors. Our previous work has shown that prenatal inhibition of the pathway produces abnormalities of brain development. In the present study kynurenine and probenecid (both 100mg/kg, doses known to increase kynurenic acid levels in the brain) were administered to female Wistar rats on embryonic days E14, E16 and E18 of gestation and the litter was allowed to develop to post-natal day P60. Western blotting revealed no changes in hippocampal expression of several proteins previously found to be altered by inhibition of the kynurenine pathway including the NMDA receptor subunits GluN1, GluN2A and GluN2B, as well as doublecortin, Proliferating Cell Nuclear Antigen (PCNA), sonic hedgehog and unco-ordinated (unc)-5H1 and 5H3. Mice lacking the enzyme kynurenine-3-monoxygenase (KMO) also showed no changes in hippocampal expression of several of these proteins or the 70-kDa and 100-kDa variants of Disrupted in Schizophrenia-1 (DISC1). Electrical excitability of pyramidal neurons in the CA1 region of hippocampal slices was unchanged, as was paired-pulse facilitation and inhibition. Long-term potentiation was decreased in the kynurenine-treated rats and in the KMO(-/-) mice, but galantamine reversed this effect in the presence of nicotinic receptor antagonists, consistent with evidence that it can potentiate glutamate at NMDA receptors. It is concluded that interference with the kynurenine pathway in utero can have lasting effects on brain function of the offspring, implying that the kynurenine pathway is involved in the regulation of early brain development.
Collapse
Affiliation(s)
- C M Forrest
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - K McNair
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - M Pisar
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - O S Khalil
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - L G Darlington
- Ashtead Hospital, The Warren, Ashtead, Surrey KT21 2SB, UK
| | - T W Stone
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
34
|
Madhavan L, Daley BF, Davidson BL, Boudreau RL, Lipton JW, Cole-Strauss A, Steece-Collier K, Collier TJ. Sonic Hedgehog Controls the Phenotypic Fate and Therapeutic Efficacy of Grafted Neural Precursor Cells in a Model of Nigrostriatal Neurodegeneration. PLoS One 2015; 10:e0137136. [PMID: 26340267 PMCID: PMC4560385 DOI: 10.1371/journal.pone.0137136] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/12/2015] [Indexed: 02/06/2023] Open
Abstract
The expression of soluble growth and survival promoting factors by neural precursor cells (NPCs) is suggested to be a prominent mechanism underlying the protective and regenerative effects of these cells after transplantation. Nevertheless, how and to what extent specific NPC-expressed factors contribute to therapeutic effects is not well understood. Using RNA silencing, the current study investigated the roles of two donor NPC molecules, namely glial cell-line derived neurotrophic factor (GDNF) and sonic hedgehog (SHH), in the protection of substantia nigra dopamine neurons in rats treated with 6-hydroxydopamine (6-OHDA). Analyses indicate that as opposed to the knock-down of GDNF, SHH inhibition caused a profound decline in nigrostriatal neuroprotection. Further, SHH silencing also curbed endogenous neurogenesis and the migration of host brdU+/dcx+ neural precursors into the striatum, which was present in the animals receiving control or GDNF silenced NPCs. A change in graft phenotype, mainly reflected by a reduced proportion of undifferentiated nestin+ cells, as well as a significantly greater host microglial activity, suggested an important role for these processes in the attenuation of neuroprotection and neurogenesis upon SHH silencing. Overall these studies reveal core mechanisms fundamental to grafted NPC-based therapeutic effects, and delineate the particular contributions of two graft-expressed molecules, SHH and GDNF, in mediating midbrain dopamine neuron protection, and host plasticity after NPC transplantation.
Collapse
Affiliation(s)
- Lalitha Madhavan
- Department of Neurology, University of Arizona, Tucson, Arizona, 85724, United States of America
- * E-mail:
| | - Brian F. Daley
- Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, 49503, United States of America
| | - Beverly L. Davidson
- Center for Cell and Molecular Therapy, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, 19104, United States of America
| | - Ryan L. Boudreau
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Jack W. Lipton
- Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, 49503, United States of America
| | - Allyson Cole-Strauss
- Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, 49503, United States of America
| | - Kathy Steece-Collier
- Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, 49503, United States of America
| | - Timothy J. Collier
- Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, 49503, United States of America
| |
Collapse
|
35
|
Giacomini A, Stagni F, Trazzi S, Guidi S, Emili M, Brigham E, Ciani E, Bartesaghi R. Inhibition of APP gamma-secretase restores Sonic Hedgehog signaling and neurogenesis in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis 2015; 82:385-396. [PMID: 26254735 PMCID: PMC4768084 DOI: 10.1016/j.nbd.2015.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/10/2015] [Accepted: 08/01/2015] [Indexed: 11/28/2022] Open
Abstract
Neurogenesis impairment starting from early developmental stages is a key determinant of intellectual disability in Down syndrome (DS). Previous evidence provided a causal relationship between neurogenesis impairment and malfunctioning of the mitogenic Sonic Hedgehog (Shh) pathway. In particular, excessive levels of AICD (amyloid precursor protein intracellular domain), a cleavage product of the trisomic gene APP (amyloid precursor protein) up-regulate transcription of Ptch1 (Patched1), the Shh receptor that keeps the pathway repressed. Since AICD results from APP cleavage by γ-secretase, the goal of the current study was to establish whether treatment with a γ-secretase inhibitor normalizes AICD levels and restores neurogenesis in trisomic neural precursor cells. We found that treatment with a selective γ-secretase inhibitor (ELND006; ELN) restores proliferation in neurospheres derived from the subventricular zone (SVZ) of the Ts65Dn mouse model of DS. This effect was accompanied by reduction of AICD and Ptch1 levels and was prevented by inhibition of the Shh pathway with cyclopamine. Treatment of Ts65Dn mice with ELN in the postnatal period P3–P15 restored neurogenesis in the SVZ and hippocampus, hippocampal granule cell number and synapse development, indicating a positive impact of treatment on brain development. In addition, in the hippocampus of treated Ts65Dn mice there was a reduction in the expression levels of various genes that are transcriptionally regulated by AICD, including APP, its origin substrate. Inhibitors of γ-secretase are currently envisaged as tools for the cure of Alzheimer's disease because they lower βamyloid levels. Current results provide novel evidence that γ-secretase inhibitors may represent a strategy for the rescue of neurogenesis defects in DS. Derangement of the mitogenic Shh pathway reduces neurogenesis in Down syndrome (DS). APP triplication causes excessive formation of its cleavage products AICD. AICD causes excessive transcription of Ptch1, the repressor of the Shh pathway. ELND006, a gamma secretase inhibitor, reduces AICD levels and Ptch1 expression. Treatment with ELND006 restores neurogenesis in the Ts65Dn mouse model of DS.
Collapse
Affiliation(s)
- Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
36
|
Expression Profile of Sonic Hedgehog Pathway Members in the Developing Human Fetal Brain. BIOMED RESEARCH INTERNATIONAL 2015; 2015:494269. [PMID: 26266257 PMCID: PMC4523658 DOI: 10.1155/2015/494269] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 03/29/2015] [Accepted: 03/30/2015] [Indexed: 11/17/2022]
Abstract
The Sonic Hedgehog (SHH) pathway plays a central role in the developing mammalian CNS. In our study, we aimed to investigate the spatiotemporal SHH pathway expression pattern in human fetal brains. We analyzed 22 normal fetal brains for Shh, Patched, Smoothened, and Gli1-3 expression by immunohistochemistry. In the telencephalon, strongest expression of Shh, Smoothened, and Gli2 was found in the cortical plate (CP) and ventricular zone. Patched was strongly upregulated in the ventricular zone and Gli1 in the CP. In the cerebellum, SHH pathway members were strongly expressed in the external granular layer (EGL). SHH pathway members significantly decreased over time in the ventricular and subventricular zone and in the cerebellar EGL, while increasing levels were found in more superficial telencephalic layers. Our findings show that SHH pathway members are strongly expressed in areas important for proliferation and differentiation and indicate a temporal expression gradient in telencephalic and cerebellar layers probably due to decreased proliferation of progenitor cells and increased differentiation. Our data about the spatiotemporal expression of SHH pathway members in the developing human brain serves as a base for the understanding of both normal and pathological CNS development.
Collapse
|
37
|
Jin Y, Raviv N, Barnett A, Bambakidis NC, Filichia E, Luo Y. The shh signaling pathway is upregulated in multiple cell types in cortical ischemia and influences the outcome of stroke in an animal model. PLoS One 2015; 10:e0124657. [PMID: 25927436 PMCID: PMC4415811 DOI: 10.1371/journal.pone.0124657] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/17/2015] [Indexed: 01/12/2023] Open
Abstract
Recently the sonic hedgehog (shh) signaling pathway has been shown to play an important role in regulating repair and regenerative responses after brain injury, including ischemia. However, the precise cellular components that express and upregulate the shh gene and the cellular components that respond to shh signaling remain to be identified. In this study, using a distal MCA occlusion model, our data show that the shh signal is upregulated both at the cortical area near the injury site and in the adjacent striatum. Multiple cell types upregulate shh signaling in ischemic brain, including neurons, reactive astrocytes and nestin-expressing cells. The shh signaling pathway genes are also expressed in the neural stem cells (NSCs) niche in the subventricular zone (SVZ). Conditional deletion of the shh gene in nestin-expressing cells both at the SVZ niche and at the ischemic site lead to significantly more severe behavioral deficits in these shh iKO mice after cortical stroke, measured using an automated open field locomotion apparatus (Student's t-test, p<0.05). In contrast, animals given post-stroke treatment with the shh signaling agonist (SAG) demonstrated less deficits in behavioral function, compared to vehicle-treated mice. At 7 days after stroke, SAG-treated mice showed higher values in multiple horizontal movement parameters compared to vehicle treated mice (Student's t-test, p<0.05) whereas there were no differences in pre-stroke measurements, (Student's t-test, p>0.05). In summary, our data demonstrate that shh signaling plays critical and ongoing roles in response to ischemic injury and modulation of shh signaling in vivo alters the functional outcome after cortical ischemic injury.
Collapse
Affiliation(s)
- Yongmin Jin
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, United States of America
| | - Nataly Raviv
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, United States of America
| | - Austin Barnett
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, United States of America
| | - Nicholas C. Bambakidis
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, United States of America
| | - Emily Filichia
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, United States of America
| | - Yu Luo
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, United States of America
- * E-mail:
| |
Collapse
|
38
|
Cuccioli V, Bueno C, Belvindrah R, Lledo PM, Martinez S. Attractive action of FGF-signaling contributes to the postnatal developing hippocampus. Hippocampus 2014; 25:486-99. [DOI: 10.1002/hipo.22386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Affiliation(s)
- V. Cuccioli
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández (CSIC-UMH); Sant Joan d'Alacant 03550 Spain
| | - C. Bueno
- Faculty of Medicine; Department of Human Anatomy and Psychobiology; IMIB-Arrixaca and CIBERSAM (Centro Investigación Biomedica en Red Salud Mental), University of Murcia E-30071; Murcia Spain
| | - R. Belvindrah
- Laboratory for Perception and Memory; Institut Pasteur; F-75015 Paris France
- Centre National de la Recherche Scientifique (CNRS); Unité Mixte de Recherche 3571 F-75015 Paris France
| | - P.-M. Lledo
- Laboratory for Perception and Memory; Institut Pasteur; F-75015 Paris France
- Centre National de la Recherche Scientifique (CNRS); Unité Mixte de Recherche 3571 F-75015 Paris France
| | - S. Martinez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández (CSIC-UMH); Sant Joan d'Alacant 03550 Spain
- Faculty of Medicine; Department of Human Anatomy and Psychobiology; IMIB-Arrixaca and CIBERSAM (Centro Investigación Biomedica en Red Salud Mental), University of Murcia E-30071; Murcia Spain
| |
Collapse
|
39
|
Nevo I, Woolard K, Cam M, Li A, Webster JD, Kotliarov Y, Kim HS, Ahn S, Walling J, Kotliarova S, Belova G, Song H, Bailey R, Zhang W, Fine HA. Identification of molecular pathways facilitating glioma cell invasion in situ. PLoS One 2014; 9:e111783. [PMID: 25365423 PMCID: PMC4218815 DOI: 10.1371/journal.pone.0111783] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/30/2014] [Indexed: 12/22/2022] Open
Abstract
Gliomas are mostly incurable secondary to their diffuse infiltrative nature. Thus, specific therapeutic targeting of invasive glioma cells is an attractive concept. As cells exit the tumor mass and infiltrate brain parenchyma, they closely interact with a changing micro-environmental landscape that sustains tumor cell invasion. In this study, we used a unique microarray profiling approach on a human glioma stem cell (GSC) xenograft model to explore gene expression changes in situ in Invading Glioma Cells (IGCs) compared to tumor core, as well as changes in host cells residing within the infiltrated microenvironment relative to the unaffected cortex. IGCs were found to have reduced expression of genes within the extracellular matrix compartment, and genes involved in cell adhesion, cell polarity and epithelial to mesenchymal transition (EMT) processes. The infiltrated microenvironment showed activation of wound repair and tissue remodeling networks. We confirmed by protein analysis the downregulation of EMT and polarity related genes such as CD44 and PARD3 in IGCs, and EFNB3, a tissue-remodeling agent enriched at the infiltrated microenvironment. OLIG2, a proliferation regulator and glioma progenitor cell marker upregulated in IGCs was found to function in enhancing migration and stemness of GSCs. Overall, our results unveiled a more comprehensive picture of the complex and dynamic cell autonomous and tumor-host interactive pathways of glioma invasion than has been previously demonstrated. This suggests targeting of multiple pathways at the junction of invading tumor and microenvironment as a viable option for glioma therapy.
Collapse
Affiliation(s)
- Ido Nevo
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kevin Woolard
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maggie Cam
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Aiguo Li
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joshua D. Webster
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yuri Kotliarov
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hong Sug Kim
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Susie Ahn
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jennifer Walling
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Svetlana Kotliarova
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Galina Belova
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hua Song
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rolanda Bailey
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wei Zhang
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Howard A. Fine
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
Pisar M, Forrest CM, Khalil OS, McNair K, Vincenten MC, Qasem S, Darlington LG, Stone TW. Modified neocortical and cerebellar protein expression and morphology in adult rats following prenatal inhibition of the kynurenine pathway. Brain Res 2014; 1576:1-17. [DOI: 10.1016/j.brainres.2014.06.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 10/25/2022]
|
41
|
Persson L, Witt RM, Galligan M, Greer PL, Eisner A, Pazyra-Murphy MF, Datta SR, Segal RA. Shh-proteoglycan interactions regulate maturation of olfactory glomerular circuitry. Dev Neurobiol 2014; 74:1255-67. [PMID: 24913191 DOI: 10.1002/dneu.22202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/30/2014] [Accepted: 06/04/2014] [Indexed: 12/27/2022]
Abstract
The olfactory system relies on precise circuitry connecting olfactory sensory neurons (OSNs) and appropriate relay and processing neurons of the olfactory bulb (OB). In mammals, the exact correspondence between specific olfactory receptor types and individual glomeruli enables a spatially precise map of glomerular activation that corresponds to distinct odors. However, the mechanisms that govern the establishment and maintenance of the glomerular circuitry are largely unknown. Here we show that high levels of Sonic Hedgehog (Shh) signaling at multiple sites enable refinement and maintenance of olfactory glomerular circuitry. Mice expressing a mutant version of Shh (Shh(Ala/Ala)), with impaired binding to proteoglycan co-receptors, exhibit disproportionately small olfactory bulbs containing fewer glomeruli. Notably, in mutant animals the correspondence between individual glomeruli and specific olfactory receptors is lost, as olfactory sensory neurons expressing different olfactory receptors converge on the same glomeruli. These deficits arise at late stages in post-natal development and continue into adulthood, indicating impaired pruning of erroneous connections within the olfactory bulb. In addition, mature Shh(Ala/Ala) mice exhibit decreased proliferation in the subventricular zone (SVZ), with particular reduction in neurogenesis of calbindin-expressing periglomerular cells. Thus, Shh interactions with proteoglycan co-receptors function at multiple locations to regulate neurogenesis and precise olfactory connectivity, thereby promoting functional neuronal circuitry.
Collapse
Affiliation(s)
- Laura Persson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ferent J, Cochard L, Faure H, Taddei M, Hahn H, Ruat M, Traiffort E. Genetic activation of Hedgehog signaling unbalances the rate of neural stem cell renewal by increasing symmetric divisions. Stem Cell Reports 2014; 3:312-23. [PMID: 25254344 PMCID: PMC4175546 DOI: 10.1016/j.stemcr.2014.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/22/2014] [Accepted: 05/22/2014] [Indexed: 02/08/2023] Open
Abstract
In the adult brain, self-renewal is essential for the persistence of neural stem cells (NSCs) throughout life, but its regulation is still poorly understood. One NSC can give birth to two NSCs or one NSC and one transient progenitor. A correct balance is necessary for the maintenance of germinal areas, and understanding the molecular mechanisms underlying NSC division mode is clearly important. Here, we report a function of the Sonic Hedgehog (SHH) receptor Patched in the direct control of long-term NSC self-renewal in the subependymal zone. We show that genetic conditional activation of SHH signaling in adult NSCs leads to their expansion and the depletion of their direct progeny. These phenotypes are associated in vitro with an increase in NSC symmetric division in a process involving NOTCH signaling. Together, our results demonstrate a tight control of adult neurogenesis and NSC renewal driven by Patched. Ptc inactivation in the adult NSCs expands the NSC pool and depletes their progeny Neurogenesis blockade is related to the increase of NSC symmetric division PTC exerts a central role via NOTCH, in the regulation of adult NSC self-renewal
Collapse
Affiliation(s)
- Julien Ferent
- Laboratory of Neurobiology and Development-CNRS, Signal Transduction and Developmental Neuropharmacology Team, Institute of Neurobiology Alfred Fessard, Gif-sur-Yvette 91198, France
| | - Loïc Cochard
- Laboratory of Neurobiology and Development-CNRS, Signal Transduction and Developmental Neuropharmacology Team, Institute of Neurobiology Alfred Fessard, Gif-sur-Yvette 91198, France
| | - Hélène Faure
- Laboratory of Neurobiology and Development-CNRS, Signal Transduction and Developmental Neuropharmacology Team, Institute of Neurobiology Alfred Fessard, Gif-sur-Yvette 91198, France
| | - Maurizio Taddei
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, 53100 Siena, Italy
| | - Heidi Hahn
- Tumor Genetics Group, Institute of Human Genetics, University Medical Center, Göttingen 37073, Germany
| | - Martial Ruat
- Laboratory of Neurobiology and Development-CNRS, Signal Transduction and Developmental Neuropharmacology Team, Institute of Neurobiology Alfred Fessard, Gif-sur-Yvette 91198, France.
| | - Elisabeth Traiffort
- Laboratory of Neurobiology and Development-CNRS, Signal Transduction and Developmental Neuropharmacology Team, Institute of Neurobiology Alfred Fessard, Gif-sur-Yvette 91198, France.
| |
Collapse
|
43
|
El Waly B, Macchi M, Cayre M, Durbec P. Oligodendrogenesis in the normal and pathological central nervous system. Front Neurosci 2014; 8:145. [PMID: 24971048 PMCID: PMC4054666 DOI: 10.3389/fnins.2014.00145] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/23/2014] [Indexed: 12/26/2022] Open
Abstract
Oligodendrocytes (OLGs) are generated late in development and myelination is thus a tardive event in the brain developmental process. It is however maintained whole life long at lower rate, and myelin sheath is crucial for proper signal transmission and neuronal survival. Unfortunately, OLGs present a high susceptibility to oxidative stress, thus demyelination often takes place secondary to diverse brain lesions or pathologies. OLGs can also be the target of immune attacks, leading to primary demyelination lesions. Following oligodendrocytic death, spontaneous remyelination may occur to a certain extent. In this review, we will mainly focus on the adult brain and on the two main sources of progenitor cells that contribute to oligodendrogenesis: parenchymal oligodendrocyte precursor cells (OPCs) and subventricular zone (SVZ)-derived progenitors. We will shortly come back on the main steps of oligodendrogenesis in the postnatal and adult brain, and summarize the key factors involved in the determination of oligodendrocytic fate. We will then shed light on the main causes of demyelination in the adult brain and present the animal models that have been developed to get insight on the demyelination/remyelination process. Finally, we will synthetize the results of studies searching for factors able to modulate spontaneous myelin repair.
Collapse
Affiliation(s)
- Bilal El Waly
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Magali Macchi
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Myriam Cayre
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Pascale Durbec
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| |
Collapse
|
44
|
Ferent J, Traiffort E. Hedgehog: Multiple Paths for Multiple Roles in Shaping the Brain and Spinal Cord. Neuroscientist 2014; 21:356-71. [PMID: 24743306 DOI: 10.1177/1073858414531457] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since the discovery of the segment polarity gene Hedgehog in Drosophila three decades ago, our knowledge of Hedgehog signaling pathway has considerably improved and paved the way to a wide field of investigations in the developing and adult central nervous system. Its peculiar transduction mechanism together with its implication in tissue patterning, neural stem cell biology, and neural tissue homeostasis make Hedgehog pathway of interest in a high number of normal or pathological contexts. Consistent with its role during brain development, misregulation of Hedgehog signaling is associated with congenital diseases and tumorigenic processes while its recruitment in damaged neural tissue may be part of the repairing process. This review focuses on the most recent data regarding the Hedgehog pathway in the developing and adult central nervous system and also its relevance as a therapeutic target in brain and spinal cord diseases.
Collapse
Affiliation(s)
- Julien Ferent
- IRCM, Molecular Biology of Neural Development, Montreal, Quebec, Canada
| | - Elisabeth Traiffort
- INSERM-Université Paris Sud, Neuroprotection and Neuroregeneration: Small Neuroactive Molecules UMR 788, Le Kremlin-Bicêtre, France
| |
Collapse
|
45
|
Abstract
As the emergence of cancer is most frequent in proliferating tissues, replication errors are considered to be at the base of this disease. This review concentrates mainly on two neural cancers, neuroblastoma and glioma, with completely different backgrounds that are well documented with respect to their ontogeny. Although clinical data on other cancers of the nervous system are available, usually little can be said about their origins. Neuroblastoma is initiated in the embryo at a moment when the nervous system (NS) is in full expansion and occasionally genomic damage can lead to neoplasia. Glioma, to the contrary, occurs in the adult brain supposed to be mostly in a postmitotic state. According to current consensus, neural stem cells located in the subventricular zone (SVZ) in the adult are thought to accumulate enough genomic mutations to diverge on a carcinogenic course leading to diverse forms of glioma. After weighing the pros and cons of this current hypothesis in this review, it will be argued that this may be improbable, yielding to the original old concept of glial origin of glioma.
Collapse
|
46
|
Khalil OS, Pisar M, Forrest CM, Vincenten MCJ, Darlington LG, Stone TW. Prenatal inhibition of the kynurenine pathway leads to structural changes in the hippocampus of adult rat offspring. Eur J Neurosci 2014; 39:1558-71. [PMID: 24646396 PMCID: PMC4368408 DOI: 10.1111/ejn.12535] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 12/13/2022]
Abstract
Glutamate receptors for N-methyl-d-aspartate (NMDA) are involved in early brain development. The kynurenine pathway of tryptophan metabolism includes the NMDA receptor agonist quinolinic acid and the antagonist kynurenic acid. We now report that prenatal inhibition of the pathway in rats with 3,4-dimethoxy-N-[4-(3-nitrophenyl)thiazol-2-yl]benzenesulphonamide (Ro61-8048) produces marked changes in hippocampal neuron morphology, spine density and the immunocytochemical localisation of developmental proteins in the offspring at postnatal day 60. Golgi–Cox silver staining revealed decreased overall numbers and lengths of CA1 basal dendrites and secondary basal dendrites, together with fewer basal dendritic spines and less overall dendritic complexity in the basal arbour. Fewer dendrites and less complexity were also noted in the dentate gyrus granule cells. More neurons containing the nuclear marker NeuN and the developmental protein sonic hedgehog were detected in the CA1 region and dentate gyrus. Staining for doublecortin revealed fewer newly generated granule cells bearing extended dendritic processes. The number of neuron terminals staining for vesicular glutamate transporter (VGLUT)-1 and VGLUT-2 was increased by Ro61-8048, with no change in expression of vesicular GABA transporter or its co-localisation with vesicle-associated membrane protein-1. These data support the view that constitutive kynurenine metabolism normally plays a role in early embryonic brain development, and that interfering with it has profound consequences for neuronal structure and morphology, lasting into adulthood.
Collapse
Affiliation(s)
- Omari S Khalil
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | | | | | | | | | | |
Collapse
|
47
|
Sawada M, Matsumoto M, Sawamoto K. Vascular regulation of adult neurogenesis under physiological and pathological conditions. Front Neurosci 2014; 8:53. [PMID: 24672424 PMCID: PMC3955849 DOI: 10.3389/fnins.2014.00053] [Citation(s) in RCA: 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: 01/08/2014] [Accepted: 02/26/2014] [Indexed: 01/16/2023] Open
Abstract
Neural stem cells in the mammalian adult brain continuously produce new neurons throughout life. Accumulating evidence in rodents suggests that various aspects of adult neurogenesis, including the genesis, migration, and maturation of new neurons, are regulated by factors derived from blood vessels and their microenvironment. Brain injury enhances both neurogenesis and angiogenesis, thereby promoting the cooperative regeneration of neurons and blood vessels. In this paper, we briefly review the mechanisms for the vascular regulation of adult neurogenesis in the ventricular-subventricular zone under physiological and pathological conditions, and discuss their clinical potential for brain regeneration strategies.
Collapse
Affiliation(s)
- Masato Sawada
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| | - Mami Matsumoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| |
Collapse
|
48
|
Ruat M, Faure H, Daynac M. Smoothened, Stem Cell Maintenance and Brain Diseases. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_83] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
49
|
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]
|
50
|
Ruan L, Lau BWM, Wang J, Huang L, Zhuge Q, Wang B, Jin K, So KF. Neurogenesis in neurological and psychiatric diseases and brain injury: from bench to bedside. Prog Neurobiol 2013; 115:116-37. [PMID: 24384539 DOI: 10.1016/j.pneurobio.2013.12.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 12/08/2013] [Accepted: 12/12/2013] [Indexed: 02/08/2023]
Abstract
Researchers who have uncovered the presence of stem cells in an adult's central nervous system have not only challenged the dogma that new neurons cannot be generated during adulthood, but also shed light on the etiology and disease mechanisms underlying many neurological and psychiatric disorders. Brain trauma, neurodegenerative diseases, and psychiatric disorders pose enormous burdens at both personal and societal levels. Although medications for these disorders are widely used, the treatment mechanisms underlying the illnesses remain largely elusive. In the past decade, an increasing amount of evidence indicate that adult neurogenesis (i.e. generating new CNS neurons during adulthood) may be involved in the pathology of different CNS disorders, and thus neurogenesis may be a potential target area for treatments. Although new neurons were shown to be a major player in mediating treatment efficacy of neurological and psychotropic drugs on cognitive functions, it is still debatable if the altered production of new neurons can cause the disorders. This review hence seeks to discuss pre and current clinical studies that demonstrate the functional impact adult neurogenesis have on neurological and psychiatric illnesses while examining the related underlying disease mechanisms.
Collapse
Affiliation(s)
- Linhui Ruan
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.
| | - Benson Wui-Man Lau
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Jixian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Lijie Huang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Brian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Kunlin Jin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.
| | - Kwok-Fai So
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China; The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China; Research Centre of Heart, Brain, Hormone and Healthy Aging, Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China; GMH Institute of CNS Regeneration, Jinan University, Guangzhou, PR China.
| |
Collapse
|