1
|
Harland AJ, Perks CM. IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis. Int J Mol Sci 2025; 26:4749. [PMID: 40429889 PMCID: PMC12111820 DOI: 10.3390/ijms26104749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Glioblastoma is a fatal and aggressive cancer with no cure. It is becoming increasingly clear that glioblastoma initiation is a result of adult neural stem cell (NSC) transformation-most likely those within the subventricular zone (SVZ). Indeed, transcriptomic analysis indicates that glioblastomas are reminiscent of a neurodevelopmental hierarchy, in which neural stem and progenitor markers are widely expressed by tumour stem-like cells. However, NSC fates and the cues that drive them are poorly understood. Studying the crosstalk within NSC niches may better inform our understanding of glioblastoma initiation and development. Insulin-like growth factor binding protein 2 (IGFBP-2) has a well-established prognostic role in glioblastoma, and cell-based mechanistic studies show the independent activation of downstream oncogenic pathways. However, IGFBP-2 is more commonly recognised as a modulator of insulin-like growth factors (IGFs) for receptor tyrosine kinase signal propagation or attenuation. In the adult human brain, both IGFBP-2 and IGF-II expression are retained in the choroid plexus (ChP) and secreted into the cerebral spinal fluid (CSF). Moreover, secretion by closely associated cells and NSCs themselves position IGFBP-2 and IGF-II as interesting factors within the NSC niche. In this review, we will highlight the experimental findings that show IGFBP-2 and IGF-II influence NSC behaviour. Moreover, we will link this to glioblastoma biology and demonstrate the requirement for further analysis of these factors in glioma stem cells (GSCs).
Collapse
Affiliation(s)
| | - Claire M. Perks
- Cancer Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK;
| |
Collapse
|
2
|
Pardo-Rodríguez B, Baraibar AM, Manero-Roig I, Luzuriaga J, Salvador-Moya J, Polo Y, Basanta-Torres R, Unda F, Mato S, Ibarretxe G, Pineda JR. Functional differentiation of human dental pulp stem cells into neuron-like cells exhibiting electrophysiological activity. Stem Cell Res Ther 2025; 16:10. [PMID: 39849603 PMCID: PMC11756023 DOI: 10.1186/s13287-025-04134-7] [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: 10/18/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND AND AIM Human dental pulp stem cells (hDPSCs) constitute a promising alternative for central nervous system (CNS) cell therapy. Unlike other human stem cells, hDPSCs can be differentiated, without genetic modification, to neural cells that secrete neuroprotective factors. However, a better understanding of their real capacity to give rise to functional neurons and integrate into synaptic networks is still needed. For that, ex vivo differentiation protocols must be refined, especially to avoid the use of fetal animal serum. The aim of our study is to improve existing differentiation protocols of hDPSCs into neuron-like cells. METHODS We compared the effects of the (1) absence or presence of fetal serum during the initial expansion phase as a step prior to switching cultures to neurodifferentiation media. We (2) improved hDPSC neurodifferentiation by adding retinoic acid (RA) and potassium chloride (KCl) pulses for 21 or 60 days and characterized the results by immunofluorescence, digital morphometric analysis, RT-qPCR and electrophysiology. RESULTS We found that neural markers like Nestin, GFAP, S100β and p75NTR were expressed differently in neurodifferentiated hDPSC cultures depending on the presence or absence of serum during the initial cell expansion phase. In addition, hDPSCs previously grown as spheroids in serum-free medium exhibited in vitro expression of neuronal markers such as doublecortin (DCX), neuronal nuclear antigen (NeuN), Ankyrin-G and MAP2 after neurodifferentiation. Presynaptic vGLUT2, Synapsin-I, and excitatory glutamatergic and inhibitory GABAergic postsynaptic scaffold proteins and receptor subunits were also present in these neurodifferentiated hDPSCs. Treatment with KCl and RA increased the amount of both voltage-gated Na+ and K+ channel subunits in neurodifferentiated hDPSCs at the transcript level. Consistently, these cells displayed voltage-dependent K+ and TTX-sensitive Na+ currents as well as spontaneous electrophysiological activity and repetitive neuronal action potentials with a full baseline potential recovery. CONCLUSION Our study demonstrates that hDPSCs can be differentiated to neuronal-like cells that display functional excitability and thus evidence the potential of these easily accessible human stem cells for nerve tissue engineering. These results highlight the importance of choosing an appropriate culture protocol to successfully neurodifferentiate hDPSCs.
Collapse
Affiliation(s)
- B Pardo-Rodríguez
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
| | - A M Baraibar
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
- Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain
- IIS Biobizkaia, Barakaldo, 48903, Spain
| | - I Manero-Roig
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
- Université de Bordeaux IINS - UMR 5297, Bordeaux, France
| | - J Luzuriaga
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
| | - J Salvador-Moya
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
| | - Y Polo
- Polimerbio SL, Donostia-San Sebastián, 20014, Spain
| | - R Basanta-Torres
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
| | - F Unda
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
| | - S Mato
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain
- Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain
- IIS Biobizkaia, Barakaldo, 48903, Spain
| | - Gaskon Ibarretxe
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain.
| | - Jose Ramon Pineda
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, 48940, Spain.
- Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.
| |
Collapse
|
3
|
Qu Y, Lim JJY, An O, Yang H, Toh YC, Chua JJE. FEZ1 participates in human embryonic brain development by modulating neuronal progenitor subpopulation specification and migrations. iScience 2023; 26:108497. [PMID: 38213789 PMCID: PMC10783620 DOI: 10.1016/j.isci.2023.108497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 09/13/2023] [Accepted: 11/17/2023] [Indexed: 01/13/2024] Open
Abstract
Mutations in the human fasciculation and elongation protein zeta 1 (FEZ1) gene are found in schizophrenia and Jacobsen syndrome patients. Here, using human cerebral organoids (hCOs), we show that FEZ1 expression is turned on early during brain development and is detectable in both neuroprogenitor subtypes and immature neurons. FEZ1 deletion disrupts expression of neuronal and synaptic development genes. Using single-cell RNA sequencing, we detected abnormal expansion of homeodomain-only protein homeobox (HOPX)- outer radial glia (oRG), concurrent with a reduction of HOPX+ oRG, in FEZ1-null hCOs. HOPX- oRGs show higher cell mobility as compared to HOPX+ oRGs. Ectopic localization of neuroprogenitors to the outer layer is seen in FEZ1-null hCOs. Anomalous encroachment of TBR2+ intermediate progenitors into CTIP2+ deep layer neurons further indicated abnormalities in cortical layer formation these hCOs. Collectively, our findings highlight the involvement of FEZ1 in early cortical brain development and how it contributes to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yinghua Qu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Jonathan Jun-Yong Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Omer An
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - John Jia En Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
- Institute for Molecular and Cell Biology, A∗STAR, Singapore 138473, Singapore
| |
Collapse
|
4
|
Vitória JJM, Trigo D, da Cruz E Silva OAB. Revisiting APP secretases: an overview on the holistic effects of retinoic acid receptor stimulation in APP processing. Cell Mol Life Sci 2022; 79:101. [PMID: 35089425 PMCID: PMC11073327 DOI: 10.1007/s00018-021-04090-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide and is characterized by the accumulation of the β-amyloid peptide (Aβ) in the brain, along with profound alterations in phosphorylation-related events and regulatory pathways. The production of the neurotoxic Aβ peptide via amyloid precursor protein (APP) proteolysis is a crucial step in AD development. APP is highly expressed in the brain and is complexly metabolized by a series of sequential secretases, commonly denoted the α-, β-, and γ-cleavages. The toxicity of resulting fragments is a direct consequence of the first cleaving event. β-secretase (BACE1) induces amyloidogenic cleavages, while α-secretases (ADAM10 and ADAM17) result in less pathological peptides. Hence this first cleavage event is a prime therapeutic target for preventing or reverting initial biochemical events involved in AD. The subsequent cleavage by γ-secretase has a reduced impact on Aβ formation but affects the peptides' aggregating capacity. An array of therapeutic strategies are being explored, among them targeting Retinoic Acid (RA) signalling, which has long been associated with neuronal health. Additionally, several studies have described altered RA levels in AD patients, reinforcing RA Receptor (RAR) signalling as a promising therapeutic strategy. In this review we provide a holistic approach focussing on the effects of isoform-specific RAR modulation with respect to APP secretases and discuss its advantages and drawbacks in subcellular AD related events.
Collapse
Affiliation(s)
- José J M Vitória
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diogo Trigo
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Odete A B da Cruz E Silva
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
5
|
LaMantia AS. Why Does the Face Predict the Brain? Neural Crest Induction, Craniofacial Morphogenesis, and Neural Circuit Development. Front Physiol 2020; 11:610970. [PMID: 33362582 PMCID: PMC7759552 DOI: 10.3389/fphys.2020.610970] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchephalic and rhombencephalic neural crest cells generate the craniofacial skeleton, special sensory organs, and subsets of cranial sensory receptor neurons. They do so while preserving the anterior-posterior (A-P) identity of their neural tube origins. This organizational principle is paralleled by central nervous system circuits that receive and process information from facial structures whose A-P identity is in register with that in the brain. Prior to morphogenesis of the face and its circuits, however, neural crest cells act as "inductive ambassadors" from distinct regions of the neural tube to induce differentiation of target craniofacial domains and establish an initial interface between the brain and face. At every site of bilateral, non-axial secondary induction, neural crest constitutes all or some of the mesenchymal compartment for non-axial mesenchymal/epithelial (M/E) interactions. Thus, for epithelial domains in the craniofacial primordia, aortic arches, limbs, the spinal cord, and the forebrain (Fb), neural crest-derived mesenchymal cells establish local sources of inductive signaling molecules that drive morphogenesis and cellular differentiation. This common mechanism for building brains, faces, limbs, and hearts, A-P axis specified, neural crest-mediated M/E induction, coordinates differentiation of distal structures, peripheral neurons that provide their sensory or autonomic innervation in some cases, and central neural circuits that regulate their behavioral functions. The essential role of this neural crest-mediated mechanism identifies it as a prime target for pathogenesis in a broad range of neurodevelopmental disorders. Thus, the face and the brain "predict" one another, and this mutual developmental relationship provides a key target for disruption by developmental pathology.
Collapse
Affiliation(s)
- Anthony-Samuel LaMantia
- Laboratory of Developmental Disorders and Genetics and Center for Neurobiology Research, Fralin Biomedical Research Institute, Department of Pediatrics, Virginia Tech-Carilion School of Medicine, Virginia Tech, Roanoke, VA, United States.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
6
|
Min HJ. Pharmacologic therapy of olfaction disroders induced by COVID-19 upper respiratory infection. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2020. [DOI: 10.5124/jkma.2020.63.9.561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Postviral olfactory disorder refers to the sensorineural olfactory loss caused by upper respiratory tract infections. With the increasing awareness of olfactory or gustatory dysfunction as a potential early symptom of coronavirus disease 2019, postviral olfactory disorder has been attracting much attention. Postviral olfactory disorder is the most common type of olfactory disorder, especially among women aged over 50 years. Systemic or topical corticosteroids have commonly been used for treating this disorder, but the mechanism of corticosteroid action is unclear. Other medical treatment options such as alpha-lipoic acids and caroverine that stimulate nerve growth factor expression or prevent glutamatergic neurotoxicity have also been used. Although these drugs have been found to be useful in the treatment of postviral olfactory disorder in previous studies, the efficacy has not been statistically proven through a meta-analysis. Recently, olfactory training has been introduced in the treatment strategy for postviral olfactory disorder. It is a safe option without side effects that can be used for treating olfactory disorders caused by upper respiratory tract infections. Further rigorous studies are needed to determine the efficacy of the combination of drug treatment and olfactory training.
Collapse
|
7
|
Prpar Mihevc S, Kokondoska Grgich V, Kopitar AN, Mohorič L, Majdič G. Neural differentiation of canine mesenchymal stem cells/multipotent mesenchymal stromal cells. BMC Vet Res 2020; 16:282. [PMID: 32778115 PMCID: PMC7418429 DOI: 10.1186/s12917-020-02493-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The ability of adipose tissue-derived multipotent mesenchymal stromal cells/mesenchymal stem cells (ASCs) to differentiate in neural lineages promises progress in the field of regenerative medicine, especially for replacing neuronal tissue damaged by different neurological disorders. Reprogramming of ASCs can be induced by the growth medium with neurogenic inductors and specific growth factors. We investigated the neural differentiation potential of canine ASCs using several growth media (KEM, NIMa, NIMb, NIMc) containing various combinations of neurogenic inductors: B27 supplement, valproic acid, forskolin, N2-supplement, and retinoic acid. Cells were first preconditioned in the pre-differentiation neural induction medium (mitogenically stimulated; STIM1), followed by the induction of neuronal differentiation. RESULTS After 3, 6, and 9 days of neural induction, elongated neural-like cells with bipolar elongations were observed, and some oval cells with light nuclei appeared. The expression of neuronal markers tubulin beta III (TUBB3), neurofilament H (NF-H), microtubule-associated protein-2 (MAP2), and glial fibrillary acidic protein (GFAP) was observed using immunocytochemistry, which confirmed the differentiation into neurons and glial cells. Flow cytometry analysis showed high GFAP expression (between 70 and 90% of all cells) after cells had been growing three days in the neural induction medium a (NIMa). Around 25% of all cells also expressed adult neuronal markers NF-H and MAP2. After nine days of ASCs differentiation, the expression of all neural markers was reduced. There were no differences between the neural differentiation of ASCs isolated from female or male dogs. CONCLUSIONS The differentiation repertoire of canine ASCs extends beyond mesodermal lineages. Using a defined neural induction medium, the canine ASCs differentiated into neural lineages and expressed markers of neuronal and glial cells, and also displayed the typical neuronal morphology. Differentiated ASCs can thus be a source of neural cellular lineages for the regenerative therapy of nerve damage and could be useful in the future for therapy or the modelling of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sonja Prpar Mihevc
- Veterinary Faculty, Institute of Preclinical Sciences, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Vesna Kokondoska Grgich
- Veterinary Faculty, Institute of Preclinical Sciences, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Andreja Nataša Kopitar
- Faculty of Medicine, Institute of Microbiology and Immunology, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia
| | - Luka Mohorič
- Animacel Ltd, Mivka 34, 1000, Ljubljana, Slovenia
| | - Gregor Majdič
- Veterinary Faculty, Institute of Preclinical Sciences, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia.
| |
Collapse
|
8
|
Cytokines Induce Monkey Neural Stem Cell Differentiation through Notch Signaling. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1308526. [PMID: 32509845 PMCID: PMC7244951 DOI: 10.1155/2020/1308526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/01/2020] [Accepted: 01/23/2020] [Indexed: 11/24/2022]
Abstract
The mammalian central nervous system (CNS) has a limited ability to renew the damaged cells after a brain or spinal cord injury whether it is nonhuman primates like monkeys or humans. Transplantation of neural stem cells (NSCs) is a potential therapy for CNS injuries due to their pluripotency and differentiation abilities. Cytokines play an important role in CNS development and repair of CNS injuries. However, the detailed cytokine signaling response in monkey neural stem cells is rarely studied. In our previous research, we isolated NSCs from the adult monkey brain and found the effects of cytokines on monkey NSCs. Now, we further analyzed the regulation mechanisms of cytokines to the proliferation of monkey NSCs such as bone morphogenic protein 4 (BMP4), BMP4/leukaemia inhibitory factor (LIF), or retinoic acid (RA)/Forskolin. The data showed that BMP4 inhibited cell proliferation to arrest, but it did not affect the stemness of NSCs. BMP4/LIF promoted the astrocyte-like differentiation of monkey NSCs, and RA/forskolin induced the neuronal differentiation of monkey NSCs. BMP4/LIF and RA/forskolin induced monkey NSC differentiation by regulating Notch signaling. These results provide some theoretical evidence for NSC therapy to brain or spinal cord injury in regenerative medicine.
Collapse
|
9
|
LaMantia A. The strengths of the genetic approach to understanding neural systems development and function: Ray Guillery's synthesis. Eur J Neurosci 2019; 49:888-899. [PMID: 29883004 PMCID: PMC6369024 DOI: 10.1111/ejn.13985] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/04/2018] [Accepted: 05/23/2018] [Indexed: 12/29/2022]
Abstract
The organization and function of sensory systems, especially the mammalian visual system, has been the focus of philosophers and scientists for centuries-from Descartes and Newton onward. Nevertheless, the utility of understanding development and its genetic foundations for deeper insight into neural function has been debated: Do you need to know how something is assembled-a car, for example-to understand how it works or how to use it-to turn on the ignition and drive? This review addresses this issue for sensory pathways. The pioneering work of the late Rainer W. (Ray) Guillery provides an unequivocal answer to this central question: Using genetics for mechanistic exploration of sensory system development yields essential knowledge of organization and function. Ray truly built the foundation for this now accepted tenet of modern neuroscience. His work on the development and reorganization of visual pathways in albino mammals-all with primary genetic mutations in genes for pigmentation-defined the genetic approach to neural systems development, function and plasticity. The work that followed his lead in a variety of sensory systems, including my own work in the developing olfactory system, proceeds directly from Ray's fundamental contributions.
Collapse
Affiliation(s)
- Anthony‐Samuel LaMantia
- Institute for Neuroscience and Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of Columbia
| |
Collapse
|
10
|
Zieger E, Garbarino G, Robert NSM, Yu JK, Croce JC, Candiani S, Schubert M. Retinoic acid signaling and neurogenic niche regulation in the developing peripheral nervous system of the cephalochordate amphioxus. Cell Mol Life Sci 2018; 75:2407-2429. [PMID: 29387904 PMCID: PMC11105557 DOI: 10.1007/s00018-017-2734-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/19/2017] [Indexed: 10/18/2022]
Abstract
The retinoic acid (RA) signaling pathway regulates axial patterning and neurogenesis in the developing central nervous system (CNS) of chordates, but little is known about its roles during peripheral nervous system (PNS) formation and about how these roles might have evolved. This study assesses the requirement of RA signaling for establishing a functional PNS in the cephalochordate amphioxus, the best available stand-in for the ancestral chordate condition. Pharmacological manipulation of RA signaling levels during embryogenesis reduces the ability of amphioxus larvae to respond to sensory stimulation and alters the number and distribution of ectodermal sensory neurons (ESNs) in a stage- and context-dependent manner. Using gene expression assays combined with immunohistochemistry, we show that this is because RA signaling specifically acts on a small population of soxb1c-expressing ESN progenitors, which form a neurogenic niche in the trunk ectoderm, to modulate ESN production during elongation of the larval body. Our findings reveal an important role for RA signaling in regulating neurogenic niche activity in the larval amphioxus PNS. Although only few studies have addressed this issue so far, comparable RA signaling functions have been reported for neurogenic niches in the CNS and in certain neurogenic placode derivatives of vertebrates. Accordingly, the here-described mechanism is likely a conserved feature of chordate embryonic and adult neural development.
Collapse
Affiliation(s)
- Elisabeth Zieger
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer, Observatoire Océanologique de Villefranche-sur-Mer, Sorbonne Universités, UPMC Université Paris 06, CNRS, 181 Chemin du Lazaret, 06230, Villefranche-sur-Mer, France
| | - Greta Garbarino
- Department of Earth, Environment and Life Sciences (Dipartimento di Scienze della Terra dell'Ambiente e della Vita, DISTAV), University of Genoa, Viale Benedetto XV 5, 16132, Genoa, Italy
| | - Nicolas S M Robert
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer, Observatoire Océanologique de Villefranche-sur-Mer, Sorbonne Universités, UPMC Université Paris 06, CNRS, 181 Chemin du Lazaret, 06230, Villefranche-sur-Mer, France
| | - Jr-Kai Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Jenifer C Croce
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer, Observatoire Océanologique de Villefranche-sur-Mer, Sorbonne Universités, UPMC Université Paris 06, CNRS, 181 Chemin du Lazaret, 06230, Villefranche-sur-Mer, France
| | - Simona Candiani
- Department of Earth, Environment and Life Sciences (Dipartimento di Scienze della Terra dell'Ambiente e della Vita, DISTAV), University of Genoa, Viale Benedetto XV 5, 16132, Genoa, Italy
| | - Michael Schubert
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer, Observatoire Océanologique de Villefranche-sur-Mer, Sorbonne Universités, UPMC Université Paris 06, CNRS, 181 Chemin du Lazaret, 06230, Villefranche-sur-Mer, France.
| |
Collapse
|
11
|
Massimino L, Flores-Garcia L, Di Stefano B, Colasante G, Icoresi-Mazzeo C, Zaghi M, Hamilton BA, Sessa A. TBR2 antagonizes retinoic acid dependent neuronal differentiation by repressing Zfp423 during corticogenesis. Dev Biol 2018; 434:231-248. [PMID: 29305158 PMCID: PMC7032051 DOI: 10.1016/j.ydbio.2017.12.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/26/2017] [Accepted: 12/28/2017] [Indexed: 01/14/2023]
Abstract
During cerebral cortex development, neural progenitors are required to elaborate a variety of cell differentiation signals to which they are continuously exposed. RA acid is a potent inducer of neuronal differentiation as it was found to influence cortical development. We report herein that TBR2, a transcription factor specific to Intermediate (Basal) Neural Progenitors (INPs), represses activation of the RA responsive element and expression of RA target genes in cell lines. This repressive action on RA signaling was functionally confirmed by the decrease of RA-mediated neuronal differentiation in neural stem cells stably overexpressing TBR2. In vivo mapping of RA activity in the developing cortex indicated that RA activity is detected in radial glial cells and subsequently downregulated in INPs, revealing a fine cell-type specific regulation of its signaling. Thus, TBR2 might be a molecular player in opposing RA signaling in INPs. Interestingly, this negative regulation is achieved at least in part by directly repressing the critical nuclear RA co-factor ZFP423. Indeed, we found ZFP423 to be expressed in the developing cortex and promote RA-dependent neuronal differentiation. These data indicate that TBR2 contributes to suppressing RA signaling in INPs, thereby enabling them to re-enter the cell cycle and delay neuronal differentiation.
Collapse
Affiliation(s)
- Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lisbeth Flores-Garcia
- Departments of Cellular&Molecular Medicine and Medicine, Moores Cancer Center, and Institute for Genomic Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0644, USA
| | - Bruno Di Stefano
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cecilia Icoresi-Mazzeo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mattia Zaghi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Bruce A Hamilton
- Departments of Cellular&Molecular Medicine and Medicine, Moores Cancer Center, and Institute for Genomic Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0644, USA
| | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| |
Collapse
|
12
|
Todd L, Suarez L, Quinn C, Fischer AJ. Retinoic Acid-Signaling Regulates the Proliferative and Neurogenic Capacity of Müller Glia-Derived Progenitor Cells in the Avian Retina. Stem Cells 2017; 36:392-405. [PMID: 29193451 DOI: 10.1002/stem.2742] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/16/2017] [Accepted: 11/03/2017] [Indexed: 12/26/2022]
Abstract
In the retina, Müller glia have the potential to become progenitor cells with the ability to proliferate and regenerate neurons. However, the ability of Müller glia-derived progenitor cells (MGPCs) to proliferate and produce neurons is limited in higher vertebrates. Using the chick model system, we investigate how retinoic acid (RA)-signaling influences the proliferation and the formation of MGPCs. We observed an upregulation of cellular RA binding proteins (CRABP) in the Müller glia of damaged retinas where the formation of MGPCs is known to occur. Activation of RA-signaling was stimulated, whereas inhibition suppressed the proliferation of MGPCs in damaged retinas and in fibroblast growth factor 2-treated undamaged retinas. Furthermore, inhibition of RA-degradation stimulated the proliferation of MGPCs. Levels of Pax6, Klf4, and cFos were upregulated in MGPCs by RA agonists and downregulated in MGPCs by RA antagonists. Activation of RA-signaling following MGPC proliferation increased the percentage of progeny that differentiated as neurons. Similarly, the combination of RA and insulin-like growth factor 1 (IGF1) significantly increased neurogenesis from retinal progenitors in the circumferential marginal zone (CMZ). In summary, RA-signaling stimulates the formation of proliferating MGPCs and enhances the neurogenic potential of MGPCs and stem cells in the CMZ. Stem Cells 2018;36:392-405.
Collapse
Affiliation(s)
- Levi Todd
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Lilianna Suarez
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Colin Quinn
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
13
|
Gkikas D, Tsampoula M, Politis PK. Nuclear receptors in neural stem/progenitor cell homeostasis. Cell Mol Life Sci 2017; 74:4097-4120. [PMID: 28638936 PMCID: PMC11107725 DOI: 10.1007/s00018-017-2571-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/06/2017] [Accepted: 06/13/2017] [Indexed: 12/13/2022]
Abstract
In the central nervous system, embryonic and adult neural stem/progenitor cells (NSCs) generate the enormous variety and huge numbers of neuronal and glial cells that provide structural and functional support in the brain and spinal cord. Over the last decades, nuclear receptors and their natural ligands have emerged as critical regulators of NSC homeostasis during embryonic development and adult life. Furthermore, substantial progress has been achieved towards elucidating the molecular mechanisms of nuclear receptors action in proliferative and differentiation capacities of NSCs. Aberrant expression or function of nuclear receptors in NSCs also contributes to the pathogenesis of various nervous system diseases. Here, we review recent advances in our understanding of the regulatory roles of steroid, non-steroid, and orphan nuclear receptors in NSC fate decisions. These studies establish nuclear receptors as key therapeutic targets in brain diseases.
Collapse
Affiliation(s)
- Dimitrios Gkikas
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece.
| |
Collapse
|
14
|
Kartal D, Yaşar M, Kartal L, Özcan I, Borlu M. Effects of isotretinoin on the olfactory function in patients with acne. An Bras Dermatol 2017; 92:191-195. [PMID: 28538877 PMCID: PMC5429103 DOI: 10.1590/abd1806-4841.20175483] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/20/2016] [Indexed: 11/22/2022] Open
Abstract
Background: Isotretinoin is a synthetic analog of vitamin A. Recent studies support a role for retinoic acid in the recovery of olfactory function following injury in mice. Objective: This study aimed at determining the effect of isotretinoin on olfactory function in patients who have acne and are otherwise healthy. Methods: Forty-five patients (aged 25-40 years) with acne were included in the study. All patients underwent a rhinological examination. Olfactory function was assessed by the Sniffin' Sticks Test. The test was assessed at baseline and in the third month of isotretinoin treatment. Results: Isotretinoin improved the performance of patients in the olfactory test. The SST score increased from 8.7±1.09 to 9.5±1.19 (p<0.001), prevalence of hyposmia decreased from 40% to 24% and normosmia increased from 60% to 75% (p=0.059). The percentage of patients whose olfactory function was categorized as "good" increased from 6% to 21.3%. This increase was statistically significant (p<0.05). Study limitations: Absence of a control group is one of the limitations of this study. Also, we did not evaluate patients with smell test after stopping isotretinoin treatment. Conclusion: We examined the effect of systemic isotretinoin on olfactory function. It can be concluded from the present investigation that isotretinoin therapy improves the sense of smell.
Collapse
Affiliation(s)
- Demet Kartal
- Department of Dermatology and Venereology - Erciyes University Faculty of Medicine - Kayseri, Turkey
| | - Mehmet Yaşar
- Department of Otorhinolaryngology - Kayseri Education and Research Hospital - Kayseri, Turkey
| | - Levent Kartal
- Department of Otorhinolaryngology - Kayseri Education and Research Hospital - Kayseri, Turkey
| | - Ibrahim Özcan
- Department of Otorhinolaryngology - Kayseri Education and Research Hospital - Kayseri, Turkey
| | - Murat Borlu
- Department of Dermatology and Venereology - Erciyes University Faculty of Medicine - Kayseri, Turkey
| |
Collapse
|
15
|
Abstract
Neural stem cells (NSCs) have been proposed as a promising cellular source for the treatment of diseases in nervous systems. NSCs can self-renew and generate major cell types of the mammalian central nervous system throughout adulthood. NSCs exist not only in the embryo, but also in the adult brain neurogenic region: the subventricular zone (SVZ) of the lateral ventricle. Embryonic stem (ES) cells acquire NSC identity with a default mechanism. Under the regulations of leukemia inhibitory factor (LIF) and fibroblast growth factors, the NSCs then become neural progenitors. Neurotrophic and differentiation factors that regulate gene expression for controlling neural cell fate and function determine the differentiation of neural progenitors in the developing mammalian brain. For clinical application of NSCs in neurodegenerative disorders and damaged neurons, there are several critical problems that remain to be resolved: 1) how to obtain enough NSCs from reliable sources for autologous transplantation; 2) how to regulate neural plasticity of different adult stem cells; 3) how to control differentiation of NSCs in the adult nervous system. In order to understand the mechanisms that control NSC differentiation and behavior, we review the ontogeny of NSCs and other stem cell plasticity of neuronal differentiation. The role of NSCs and their regulation by neurotrophic factors in CNS development are also reviewed.
Collapse
Affiliation(s)
- Yi-Chao Hsu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Don-Ching Lee
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Ing-Ming Chiu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
- Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Institute of Medical Technology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
16
|
Kim JY, Shaker MR, Lee JH, Lee B, Kim H, Sun W. Identification of molecular markers distinguishing adult neural stem cells in the subventricular and subcallosal zones. Anim Cells Syst (Seoul) 2017; 21:152-159. [PMID: 30460064 PMCID: PMC6138335 DOI: 10.1080/19768354.2017.1324522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells (NSCs) in the adult subventricular zone (SVZ) are regionally specified and have distinct molecular gene expression signatures. Recently, we identified the subcallosal zone (SCZ) as a novel brain region where adult NSCs maintain and spontaneously produce neuroblasts. In an attempt to isolate genes specifically expressed in the SCZ or SVZ, microarray analyses of their differentially expressing transcripts were done. The comparison between neurospheres generated from SVZ and SCZ revealed differential expression >1.5-fold in two groups in only 83 genes, representing <0.03% of the genes examined, suggesting that these two populations are largely similar. The differential expression patterns SCZ and SVZ genes were confirmed by RT-PCR and Western blots. The selective expressions of two genes (CRBP1, HMGA1) in SVZ-NSCs were further confirmed by immunohistochemistry. These molecular markers could be useful for further molecular and cellular characterization of NSCs.
Collapse
Affiliation(s)
- Joo Yeon Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Mohammed R. Shaker
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Ju-Hyun Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Boram Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Hyun Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
17
|
New Insights Into the Roles of Retinoic Acid Signaling in Nervous System Development and the Establishment of Neurotransmitter Systems. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 330:1-84. [PMID: 28215529 DOI: 10.1016/bs.ircmb.2016.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Secreted chiefly from the underlying mesoderm, the morphogen retinoic acid (RA) is well known to contribute to the specification, patterning, and differentiation of neural progenitors in the developing vertebrate nervous system. Furthermore, RA influences the subtype identity and neurotransmitter phenotype of subsets of maturing neurons, although relatively little is known about how these functions are mediated. This review provides a comprehensive overview of the roles played by RA signaling during the formation of the central and peripheral nervous systems of vertebrates and highlights its effects on the differentiation of several neurotransmitter systems. In addition, the evolutionary history of the RA signaling system is discussed, revealing both conserved properties and alternate modes of RA action. It is proposed that comparative approaches should be employed systematically to expand our knowledge of the context-dependent cellular mechanisms controlled by the multifunctional signaling molecule RA.
Collapse
|
18
|
Chau KF, Springel MW, Broadbelt KG, Park HY, Topal S, Lun MP, Mullan H, Maynard T, Steen H, LaMantia AS, Lehtinen MK. Progressive Differentiation and Instructive Capacities of Amniotic Fluid and Cerebrospinal Fluid Proteomes following Neural Tube Closure. Dev Cell 2016; 35:789-802. [PMID: 26702835 DOI: 10.1016/j.devcel.2015.11.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 09/28/2015] [Accepted: 11/16/2015] [Indexed: 01/04/2023]
Abstract
After neural tube closure, amniotic fluid (AF) captured inside the neural tube forms the nascent cerebrospinal fluid (CSF). Neuroepithelial stem cells contact CSF-filled ventricles, proliferate, and differentiate to form the mammalian brain, while neurogenic placodes, which generate cranial sensory neurons, remain in contact with the AF. Using in vivo ultrasound imaging, we quantified the expansion of the embryonic ventricular-CSF space from its inception. We developed tools to obtain pure AF and nascent CSF, before and after neural tube closure, and to define how the AF and CSF proteomes diverge during mouse development. Using embryonic neural explants, we demonstrate that age-matched fluids promote Sox2-positive neurogenic identity in developing forebrain and olfactory epithelia. Nascent CSF also stimulates SOX2-positive self-renewal of forebrain progenitor cells, some of which is attributable to LIFR signaling. Our Resource should facilitate the investigation of fluid-tissue interactions during this highly vulnerable stage of early brain development.
Collapse
Affiliation(s)
- Kevin F Chau
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Mark W Springel
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kevin G Broadbelt
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Hye-Yeon Park
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Salih Topal
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Melody P Lun
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hillary Mullan
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Thomas Maynard
- Department of Pharmacology and Physiology, Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Hanno Steen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anthony S LaMantia
- Department of Pharmacology and Physiology, Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Souza CS, Paulsen BS, Devalle S, Lima Costa S, Borges HL, Rehen SK. Commitment of human pluripotent stem cells to a neural lineage is induced by the pro-estrogenic flavonoid apigenin. ACTA ACUST UNITED AC 2015. [DOI: 10.3402/arb.v2.29244] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
20
|
LaMantia AS, Moody SA, Maynard TM, Karpinski BA, Zohn IE, Mendelowitz D, Lee NH, Popratiloff A. Hard to swallow: Developmental biological insights into pediatric dysphagia. Dev Biol 2015; 409:329-42. [PMID: 26554723 DOI: 10.1016/j.ydbio.2015.09.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/10/2015] [Accepted: 09/15/2015] [Indexed: 12/16/2022]
Abstract
Pediatric dysphagia-feeding and swallowing difficulties that begin at birth, last throughout childhood, and continue into maturity--is one of the most common, least understood complications in children with developmental disorders. We argue that a major cause of pediatric dysphagia is altered hindbrain patterning during pre-natal development. Such changes can compromise craniofacial structures including oropharyngeal muscles and skeletal elements as well as motor and sensory circuits necessary for normal feeding and swallowing. Animal models of developmental disorders that include pediatric dysphagia in their phenotypic spectrum can provide mechanistic insight into pathogenesis of feeding and swallowing difficulties. A fairly common human genetic developmental disorder, DiGeorge/22q11.2 Deletion Syndrome (22q11DS) includes a substantial incidence of pediatric dysphagia in its phenotypic spectrum. Infant mice carrying a parallel deletion to 22q11DS patients have feeding and swallowing difficulties that approximate those seen in pediatric dysphagia. Altered hindbrain patterning, craniofacial malformations, and changes in cranial nerve growth prefigure these difficulties. Thus, in addition to craniofacial and pharyngeal anomalies that arise independently of altered neural development, pediatric dysphagia may result from disrupted hindbrain patterning and its impact on peripheral and central neural circuit development critical for feeding and swallowing. The mechanisms that disrupt hindbrain patterning and circuitry may provide a foundation to develop novel therapeutic approaches for improved clinical management of pediatric dysphagia.
Collapse
Affiliation(s)
- Anthony-Samuel LaMantia
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Sally A Moody
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Thomas M Maynard
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Beverly A Karpinski
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Irene E Zohn
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Center for Neuroscience Research, Children's National Health System, Washington D.C., USA
| | - David Mendelowitz
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Norman H Lee
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Anastas Popratiloff
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| |
Collapse
|
21
|
Current Neurogenic and Neuroprotective Strategies to Prevent and Treat Neurodegenerative and Neuropsychiatric Disorders. Neuromolecular Med 2015; 17:404-22. [PMID: 26374113 DOI: 10.1007/s12017-015-8369-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/22/2015] [Indexed: 12/31/2022]
Abstract
The adult central nervous system is commonly known to have a very limited regenerative capacity. The presence of functional stem cells in the brain can therefore be seen as a paradox, since in other organs these are known to counterbalance cell loss derived from pathological conditions. This fact has therefore raised the possibility to stimulate neural stem cell differentiation and proliferation or survival by either stem cell replacement therapy or direct administration of neurotrophic factors or other proneurogenic molecules, which in turn has also originated regenerative medicine for the treatment of otherwise incurable neurodegenerative and neuropsychiatric disorders that take a huge toll on society. This may be facilitated by the fact that many of these disorders converge on similar pathophysiological pathways: excitotoxicity, oxidative stress, neuroinflammation, mitochondrial failure, excessive intracellular calcium and apoptosis. This review will therefore focus on the most promising achievements in promoting neuroprotection and neuroregeneration reported to date.
Collapse
|
22
|
Gorris R, Fischer J, Erwes KL, Kesavan J, Peterson DA, Alexander M, Nöthen MM, Peitz M, Quandel T, Karus M, Brüstle O. Pluripotent stem cell-derived radial glia-like cells as stable intermediate for efficient generation of human oligodendrocytes. Glia 2015; 63:2152-67. [PMID: 26123132 DOI: 10.1002/glia.22882] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 11/10/2022]
Abstract
Neural precursor cells (NPCs) derived from human pluripotent stem cells (hPSCs) represent an attractive tool for the in vitro generation of various neural cell types. However, the developmentally early NPCs emerging during hPSC differentiation typically show a strong propensity for neuronal differentiation, with more limited potential for generating astrocytes and, in particular, for generating oligodendrocytes. This phenomenon corresponds well to the consecutive and protracted generation of neurons and GLIA during normal human development. To obtain a more gliogenic NPC type, we combined growth factor-mediated expansion with pre-exposure to the differentiation-inducing agent retinoic acid and subsequent immunoisolation of CD133-positive cells. This protocol yields an adherent and self-renewing population of hindbrain/spinal cord radial glia (RG)-like neural precursor cells (RGL-NPCs) expressing typical neural stem cell markers such as nestin, ASCL1, SOX2, and PAX6 as well as RG markers BLBP, GLAST, vimentin, and GFAP. While RGL-NPCs maintain the ability for tripotential differentiation into neurons, astrocytes, and oligodendrocytes, they exhibit greatly enhanced propensity for oligodendrocyte generation. Under defined differentiation conditions promoting the expression of the major oligodendrocyte fate-determinants OLIG1/2, NKX6.2, NKX2.2, and SOX10, RGL-NPCs efficiently convert into NG2-positive oligodendroglial progenitor cells (OPCs) and are subsequently capable of in vivo myelination. Representing a stable intermediate between PSCs and OPCs, RGL-NPCs expedite the generation of PSC-derived oligodendrocytes with O4-, 4860-, and myelin basic protein (MBP)-positive cells that already appear within 7 weeks following growth factor withdrawal-induced differentiation. Thus, RGL-NPCs may serve as robust tool for time-efficient generation of human oligodendrocytes from embryonic and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Raphaela Gorris
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Julia Fischer
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Kim Lina Erwes
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Jaideep Kesavan
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Daniel A Peterson
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany.,Center for Stem Cell and Regenerative Medicine, Department of Neuroscience, Rosalind Franklin University of Medicine and Science, Chicago, Illinois
| | - Michael Alexander
- Institute of Human Genetics, LIFE & BRAIN Center, University of Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, LIFE & BRAIN Center, University of Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany.,German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Tamara Quandel
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Michael Karus
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| |
Collapse
|
23
|
Chang JT, Lehtinen MK, Sive H. Zebrafish cerebrospinal fluid mediates cell survival through a retinoid signaling pathway. Dev Neurobiol 2015; 76:75-92. [PMID: 25980532 PMCID: PMC4644717 DOI: 10.1002/dneu.22300] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 11/07/2022]
Abstract
Cerebrospinal fluid (CSF) includes conserved factors whose function is largely unexplored. To assess the role of CSF during embryonic development, CSF was repeatedly drained from embryonic zebrafish brain ventricles soon after their inflation. Removal of CSF increased cell death in the diencephalon, indicating a survival function. Factors within the CSF are required for neuroepithelial cell survival as injected mouse CSF but not artificial CSF could prevent cell death after CSF depletion. Mass spectrometry analysis of the CSF identified retinol binding protein 4 (Rbp4), which transports retinol, the precursor to retinoic acid (RA). Consistent with a role for Rbp4 in cell survival, inhibition of Rbp4 or RA synthesis increased neuroepithelial cell death. Conversely, ventricle injection of exogenous human RBP4 plus retinol, or RA alone prevented cell death after CSF depletion. Zebrafish rbp4 is highly expressed in the yolk syncytial layer, suggesting Rbp4 protein and retinol/RA precursors can be transported into the CSF from the yolk. In accord with this suggestion, injection of human RBP4 protein into the yolk prevents neuroepithelial cell death in rbp4 loss‐of‐function embryos. Together, these data support the model that Rbp4 and RA precursors are present within the CSF and used for synthesis of RA, which promotes embryonic neuroepithelial survival. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 75–92, 2016
Collapse
Affiliation(s)
- Jessica T Chang
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, Massachusetts, 02142.,Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, 02115
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, Massachusetts, 02142.,Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| |
Collapse
|
24
|
Jin W, Xu YP, Yang AH, Xing YQ. In vitro induction and differentiation of umbilical cord mesenchymal stem cells into neuron-like cells by all-trans retinoic acid. Int J Ophthalmol 2015; 8:250-6. [PMID: 25938036 DOI: 10.3980/j.issn.2222-3959.2015.02.07] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/06/2014] [Indexed: 12/29/2022] Open
Abstract
AIM To determine the optimal concentration for inducing the differentiation of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) into neuron-like cells, although it is understood that all-trans retinoic acid (ATRA) regulates cell proliferation in the nervous system by modulating the balance between mitosis and apoptosis. METHODS The abilities of ATRA to promote apoptosis as well as neural differentiation were assessed in cultured hUC-MSCs by morphological observation, MTT assay, annexin V-FITC/PI flow cytometry and immunocytochemistry. RESULTS The data showed that low concentrations of ATRA (0.5 µmol, 0.25 µmol) had no effect on the number of cells. However, treatment with 1.0 µmol or 2.0 µmol ATRA induced a 24.16% and 52.67% reduction in cell number, respectively, compared with vehicle-treated cultures. Further, 4.0 µmol ATRA had a potent effect on cell number, with almost no adherent cells recovered after 24h. We further showed that 0.5 µmol ATRA caused these cells to express characteristic markers of neuronal progenitor cells. CONCLUSION Taken together, we conclude that ATRA has a dose-dependent influence on the neural differentiation and apoptosis of hUC-MSCs. These findings have implications on the use of ATRA-differentiated hUC-MSCs for the study of neural degeneration diseases.
Collapse
Affiliation(s)
- Wei Jin
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yao-Peng Xu
- Department of Urology, Wuhan General Hospital of Guangzhou Military Command, Wuhan 430072, Hubei Province, China
| | - An-Huai Yang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yi-Qiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| |
Collapse
|
25
|
Choschzick I, Hirseland E, Cramer H, Schultz S, Leppert J, Tronnier V, Zechel C. Responsiveness of stem-like human glioma cells to all-trans retinoic acid and requirement of retinoic acid receptor isotypes α, β and γ. Neuroscience 2014; 279:44-64. [PMID: 25171789 DOI: 10.1016/j.neuroscience.2014.07.078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/01/2014] [Accepted: 07/30/2014] [Indexed: 01/12/2023]
Abstract
Retinoic acid (RA) is required for development and homeostasis of the normal mammalian brain and may play a role in the initiation and progression of malignant brain tumors, such as the glioblastoma multiforme (GBM) and the gliosarcoma (Gsarc). The subpopulation of stem-like glioma cells (SLGCs) was shown to resist standard glioma radio-/chemotherapy and to propagate tumor regrowth. We used phenotypically distinct, self-renewing SLGC lines from six human GBMs, two Gsarcs, and two subcloned SLGC derivatives in order to investigate their responsiveness to all-trans retinoic acid (atRA) and to identify the RA-receptor (RAR) isotypes involved. In general, atRA exerted a pro-proliferative and pro-survival effect on SLGCs, though the efficacy was distinct. By means of RAR isotype-selective retinoids we disclosed that these effects were mediated by RARα and RARγ, except for one SLGC line, in which the pro-proliferative signal was induced by the RARβ-selective retinoid. Only one GBM-derived cell line (T1338) and a subpopulation of another (T1389) displayed neural differentiation in response to atRA. Differentiation of T1338 was induced by RARα and RARγ isotype-selective retinoids, associated with down-regulation of Sox2, and the failure to induce orthotopic tumors in the brains of SCID mice. The differential responsiveness of the SLGC lines appeared unrelated to the expression of RARβ, as (i) atRA augmented RAR isotype mRNA expression and particularly rarβ mRNA in all SLGC lines, (ii) rarβ promoter hypomethylation in the SLGC lines was not related to differentiation and (iii) the induction of T1338 differentiation was by RARα- and RARγ-selective ligands.
Collapse
Affiliation(s)
- I Choschzick
- Department of Neurosurgery, University of Lübeck, D-23538 Lübeck, Germany
| | - E Hirseland
- Department of Neurosurgery, University of Lübeck, D-23538 Lübeck, Germany; Department of Radiation Oncology, University of Lübeck, D-23538 Lübeck, Germany
| | - H Cramer
- Department of Neurosurgery, University of Lübeck, D-23538 Lübeck, Germany
| | - S Schultz
- Department of Neurosurgery, University of Lübeck, D-23538 Lübeck, Germany; Department of Radiation Oncology, University of Lübeck, D-23538 Lübeck, Germany
| | - J Leppert
- Department of Neurosurgery, University of Lübeck, D-23538 Lübeck, Germany
| | - V Tronnier
- Department of Neurosurgery, University of Lübeck, D-23538 Lübeck, Germany
| | - C Zechel
- Department of Neurosurgery, University of Lübeck, D-23538 Lübeck, Germany.
| |
Collapse
|
26
|
Abstract
Retinoids are essential in the development and function of several organs, exerting potent effects on stem cell systems. All-trans retinoic acid, through binding to the retinoic acid response elements, alters transcription of numerous genes in stem cells, leading to an exit from the self-renewing state and promoting differentiation. In the kidney, retinoids protect against injury and ameliorate function in multiple experimental models of disease. Recent evidence suggests that retinoids act on renal progenitors by promoting their differentiation into mature podocytes and retinoic acid-induced podocyte differentiation is impaired by proteinuria because of sequestration of retinoic acid by albumin. However, retinoic acid administration can revert renal progenitor differentiation and promote podocyte regeneration. A more complete understanding of retinoid-dependent renal progenitor differentiation into podocytes should reward us with new insights into the mechanisms of progression toward glomerulosclerosis.
Collapse
Affiliation(s)
- Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the Development of de novo Therapies, University of Florence, Florence, Italy.
| | - Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the Development of de novo Therapies, University of Florence, Florence, Italy; Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | - Laura Lasagni
- Excellence Centre for Research, Transfer and High Education for the Development of de novo Therapies, University of Florence, Florence, Italy
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of de novo Therapies, University of Florence, Florence, Italy; Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy; Pediatric Nephrology Unit, Meyer Children's University Hospital, Florence, Italy
| |
Collapse
|
27
|
Retinoic Acid, under Cerebrospinal Fluid Control, Induces Neurogenesis during Early Brain Development. J Dev Biol 2014. [DOI: 10.3390/jdb2020072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
28
|
Kanski R, van Strien ME, van Tijn P, Hol EM. A star is born: new insights into the mechanism of astrogenesis. Cell Mol Life Sci 2014; 71:433-47. [PMID: 23907612 PMCID: PMC11113452 DOI: 10.1007/s00018-013-1435-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/10/2013] [Accepted: 07/22/2013] [Indexed: 12/22/2022]
Abstract
Astrocytes emerge as crucial cells for proper neuronal functioning in the developing and adult brain. Neurons and astrocytes are sequentially generated from the same pool of neural stem cells (NSCs). Tight regulation of the neuron-to-astrocyte switch is critical for (1) the generation of a balanced number of astrocytes and neurons and (2) neuronal circuit formation, since newborn astrocytes regulate synapse formation. This review focuses on signaling pathways that instruct astrogenesis, incorporating recently discovered intrinsic and extrinsic regulators. The canonical pathway of astrocytic gene expression, JAK/STAT signaling, is inhibited during neurogenesis to prevent premature astrocyte differentiation. At the onset of astrogenesis, Notch signaling induces epigenetic remodeling of astrocytic genes like glial fibrillary acidic protein to change NSC competence. In turn, astrogenesis is initiated by signals received from newborn neurons. We highlight how key molecular pathways like JAK/STAT and Notch are integrated in a complex network of environmental signals and epigenetic and transcriptional regulators to determine NSC differentiation. It is essential to understand NSC differentiation in respect to future NSC-based therapies for brain diseases, as transplanted NSCs preferentially become astrocytes. As emphasized in this review, many clues in this respect can be learned from development.
Collapse
Affiliation(s)
- Regina Kanski
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Miriam E. van Strien
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Paula van Tijn
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Hubrecht Institute, an Institute of the Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
| | - Elly M. Hol
- Astrocyte Biology and Neurodegeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Oyarce K, Bongarzone ER, Nualart F. Unconventional Neurogenic Niches and Neurogenesis Modulation by Vitamins. ACTA ACUST UNITED AC 2014. [PMID: 26203401 DOI: 10.4172/2157-7633.1000184] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although the generation of new neurons occurs in adult mammals, it has been classically described in two defined regions of the brain denominated neurogenic niches: the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus. In these regions, neural stem cells give rise to new neurons and glia, which functionally integrate into the existing circuits under physiological conditions. However, accumulating evidence indicates the presence of neurogenic potential in other brain regions, from which multipotent precursors can be isolated and differentiated in vitro. In some of these regions, neuron generation occurs at low levels; however, the addition of growth factors, hormones or other signaling molecules increases the proliferation and differentiation of precursor cells. In addition, vitamins, which are micronutrients necessary for normal brain development, and whose deficiency produces neurological impairments, have a regulatory effect on neural stem cells in vitro and in vivo. In the present review, we will describe the progress that has been achieved in determining the neurogenic potential in other regions, known as unconventional niches, as well as the characteristics of the neural stem cells described for each region. Finally, we will revisit the roles of commonly known vitamins as modulators of precursor cell proliferation and differentiation, and their role in the complex and tight molecular signaling that impacts these neurogenic niches.
Collapse
Affiliation(s)
- Karina Oyarce
- Laboratory of Neurobiology and Stem Cells, Center for Advanced Microscopy CMA BIO BIO, Concepcion University, Concepción, Chile
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, USA
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells, Center for Advanced Microscopy CMA BIO BIO, Concepcion University, Concepción, Chile
| |
Collapse
|
30
|
The choroid plexus and cerebrospinal fluid: emerging roles in development, disease, and therapy. J Neurosci 2013; 33:17553-9. [PMID: 24198345 DOI: 10.1523/jneurosci.3258-13.2013] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although universally recognized as the source of cerebrospinal fluid (CSF), the choroid plexus (ChP) has been one of the most understudied tissues in neuroscience. The reasons for this are multiple and varied, including historical perceptions about passive and permissive roles for the ChP, experimental issues, and lack of clinical salience. However, recent work on the ChP and instructive signals in the CSF have sparked new hypotheses about how the ChP and CSF provide unexpected means for regulating nervous system structure and function in health and disease, as well as new ChP-based therapeutic approaches using pluripotent stem cell technology. This minisymposium combines new and established investigators to capture some of the newfound excitement surrounding the ChP-CSF system.
Collapse
|
31
|
Meechan DW, Rutz HLH, Fralish MS, Maynard TM, Rothblat LA, LaMantia AS. Cognitive ability is associated with altered medial frontal cortical circuits in the LgDel mouse model of 22q11.2DS. Cereb Cortex 2013; 25:1143-51. [PMID: 24217989 DOI: 10.1093/cercor/bht308] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We established a relationship between cognitive deficits and cortical circuits in the LgDel model of 22q11 Deletion Syndrome (22q11DS)-a genetic syndrome with one of the most significant risks for schizophrenia and autism. In the LgDel mouse, optimal acquisition, execution, and reversal of a visually guided discrimination task, comparable to executive function tasks in primates including humans, are compromised; however, there is significant individual variation in degree of impairment. The task relies critically on the integrity of circuits in medial anterior frontal cortical regions. Accordingly, we analyzed neuronal changes that reflect previously defined 22q11DS-related alterations of cortical development in the medial anterior frontal cortex of the behaviorally characterized LgDel mice. Interneuron placement, synapse distribution, and projection neuron frequency are altered in this region. The magnitude of one of these changes, layer 2/3 projection neuron frequency, is a robust predictor of behavioral performance: it is substantially and selectively lower in animals with the most significant behavioral deficits. These results parallel correlations of volume reduction and altered connectivity in comparable cortical regions with diminished executive function in 22q11DS patients. Apparently, 22q11 deletion alters behaviorally relevant circuits in a distinct cortical region that are essential for cognitive function.
Collapse
Affiliation(s)
- D W Meechan
- Department of Pharmacology and Physiology GW Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA
| | - H L H Rutz
- Department of Psychology GW Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA
| | - M S Fralish
- Department of Pharmacology and Physiology GW Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA
| | - T M Maynard
- Department of Pharmacology and Physiology GW Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA
| | - L A Rothblat
- Department of Psychology GW Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA
| | - A-S LaMantia
- Department of Pharmacology and Physiology GW Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
32
|
Micucci JA, Layman WS, Hurd EA, Sperry ED, Frank SF, Durham MA, Swiderski DL, Skidmore JM, Scacheri PC, Raphael Y, Martin DM. CHD7 and retinoic acid signaling cooperate to regulate neural stem cell and inner ear development in mouse models of CHARGE syndrome. Hum Mol Genet 2013; 23:434-48. [PMID: 24026680 DOI: 10.1093/hmg/ddt435] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
CHARGE syndrome is a multiple congenital anomaly disorder that leads to life-threatening birth defects, such as choanal atresia and cardiac malformations as well as multiple sensory impairments, that affect hearing, vision, olfaction and balance. CHARGE is caused by heterozygous mutations in CHD7, which encodes an ATP-dependent chromatin remodeling enzyme. Identification of the mechanisms underlying neurological and sensory defects in CHARGE is a first step toward developing treatments for CHARGE individuals. Here, we used mouse models of Chd7 deficiency to explore the function of CHD7 in the development of the subventricular zone (SVZ) neural stem cell niche and inner ear, structures that are important for olfactory bulb neurogenesis and hearing and balance, respectively. We found that loss of Chd7 results in cell-autonomous proliferative, neurogenic and self-renewal defects in the perinatal and mature mouse SVZ stem cell niche. Modulation of retinoic acid (RA) signaling prevented in vivo inner ear and in vitro neural stem cell defects caused by Chd7 deficiency. Our findings demonstrate critical, cooperative roles for RA and CHD7 in SVZ neural stem cell function and inner ear development, suggesting that altered RA signaling may be an effective method for treating Chd7 deficiency.
Collapse
|
33
|
Orsolits B, Borsy A, Madarász E, Mészáros Z, Kőhidi T, Markó K, Jelitai M, Welker E, Környei Z. Retinoid machinery in distinct neural stem cell populations with different retinoid responsiveness. Stem Cells Dev 2013; 22:2777-93. [PMID: 23734950 DOI: 10.1089/scd.2012.0422] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Retinoic acid (RA) is present at sites of neurogenesis in both the embryonic and adult brain. While it is widely accepted that RA signaling is involved in the regulation of neural stem cell differentiation, little is known about vitamin A utilization and biosynthesis of active retinoids in the neurogenic niches, or about the details of retinoid metabolism in neural stem cells and differentiating progenies. Here we provide data on retinoid responsiveness and RA production of distinct neural stem cell/neural progenitor populations. In addition, we demonstrate differentiation-related changes in the expression of genes encoding proteins of the retinoid machinery, including components responsible for uptake (Stra6) and storage (Lrat) of vitamin A, transport of retinoids (Rbp4, CrbpI, CrabpI-II), synthesis (Rdh10, Raldh1-4), degradation of RA (Cyp26a1-c1) and RA signaling (Rarα,β,γ, Rxrα,β,γ). We show that both early embryonic neuroectodermal (NE-4C) stem cells and late embryonic or adult derived radial glia like progenitors (RGl cells) are capable to produce bioactive retinoids but respond differently to retinoid signals. However, while neuronal differentiation of RGl cells can not be induced by RA, neuron formation by NE-4C cells is initiated by both RA and RA-precursors (retinol or retinyl acetate). The data indicate that endogenous RA production, at least in some neural stem cell populations, may result in autocrine regulation of neuronal differentiation.
Collapse
Affiliation(s)
- Barbara Orsolits
- 1 Institute of Experimental Medicine , Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Butler Tjaden NE, Trainor PA. The developmental etiology and pathogenesis of Hirschsprung disease. Transl Res 2013; 162:1-15. [PMID: 23528997 PMCID: PMC3691347 DOI: 10.1016/j.trsl.2013.03.001] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 02/25/2013] [Accepted: 03/01/2013] [Indexed: 02/08/2023]
Abstract
The enteric nervous system is the part of the autonomic nervous system that directly controls the gastrointestinal tract. Derived from a multipotent, migratory cell population called the neural crest, a complete enteric nervous system is necessary for proper gut function. Disorders that arise as a consequence of defective neural crest cell development are termed neurocristopathies. One such disorder is Hirschsprung disease (HSCR), also known as congenital megacolon or intestinal aganglionosis. HSCR occurs in 1/5000 live births and typically presents with the inability to pass meconium, along with abdominal distension and discomfort that usually requires surgical resection of the aganglionic bowel. This disorder is characterized by a congenital absence of neurons in a portion of the intestinal tract, usually the distal colon, because of a disruption of normal neural crest cell migration, proliferation, differentiation, survival, and/or apoptosis. The inheritance of HSCR disease is complex, often non-Mendelian, and characterized by variable penetrance. Extensive research has identified a number of key genes that regulate neural crest cell development in the pathogenesis of HSCR including RET, GDNF, GFRα1, NRTN, EDNRB, ET3, ZFHX1B, PHOX2b, SOX10, and SHH. However, mutations in these genes account for only ∼50% of the known cases of HSCR. Thus, other genetic mutations and combinations of genetic mutations and modifiers likely contribute to the etiology and pathogenesis of HSCR. The aims of this review are to summarize the HSCR phenotype, diagnosis, and treatment options; to discuss the major genetic causes and the mechanisms by which they disrupt normal enteric neural crest cell development; and to explore new pathways that may contribute to HSCR pathogenesis.
Collapse
|
35
|
Stergiopoulos A, Politis PK. The role of nuclear receptors in controlling the fine balance between proliferation and differentiation of neural stem cells. Arch Biochem Biophys 2013; 534:27-37. [DOI: 10.1016/j.abb.2012.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 08/23/2012] [Accepted: 09/20/2012] [Indexed: 12/22/2022]
|
36
|
Pro- and anti-mitogenic actions of pituitary adenylate cyclase-activating polypeptide in developing cerebral cortex: potential mediation by developmental switch of PAC1 receptor mRNA isoforms. J Neurosci 2013; 33:3865-78. [PMID: 23447598 DOI: 10.1523/jneurosci.1062-12.2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During corticogenesis, pituitary adenylate cyclase-activating polypeptide (PACAP; ADCYAP1) may contribute to proliferation control by activating PAC1 receptors of neural precursors in the embryonic ventricular zone. PAC1 receptors, specifically the hop and short isoforms, couple differentially to and activate distinct pathways that produce pro- or anti-mitogenic actions. Previously, we found that PACAP was an anti-mitogenic signal from embryonic day 13.5 (E13.5) onward both in culture and in vivo and activated cAMP signaling through the short isoform. However, we now find that mice deficient in PACAP exhibited a decrease in the BrdU labeling index (LI) in E9.5 cortex, suggesting that PACAP normally promotes proliferation at this stage. To further define mechanisms, we established a novel culture model in which the viability of very early cortical precursors (E9.5 mouse and E10.5 rat) could be maintained. At this stage, we found that PACAP evoked intracellular calcium fluxes and increased phospho-PKC levels, as well as stimulated G1 cyclin mRNAs and proteins, S-phase entry, and proliferation without affecting cell survival. Significantly, expression of hop receptor isoform was 24-fold greater than the short isoform at E10.5, a ratio that was reversed at E14.5 when short expression was 15-fold greater and PACAP inhibited mitogenesis. Enhanced hop isoform expression, elicited by in vitro treatment of E10.5 precursors with retinoic acid, correlated with sustained pro-mitogenic action of PACAP beyond the developmental switch. Conversely, depletion of hop receptor using short-hairpin RNA abolished PACAP mitogenic stimulation at E10.5. These observations suggest that PACAP elicits temporally specific effects on cortical proliferation via developmentally regulated expression of specific receptor isoforms.
Collapse
|
37
|
Carnicero E, Alonso M, Carretero R, Lamus F, Moro J, de la Mano A, Fernández J, Gato A. Embryonic Cerebrospinal Fluid Activates Neurogenesis of Neural Precursors within the Subventricular Zone of the Adult Mouse Brain. Cells Tissues Organs 2013; 198:398-404. [DOI: 10.1159/000356983] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2013] [Indexed: 11/19/2022] Open
|
38
|
HONG JIANGCONG, WU GUANGWEN, ZOU YULONG, TAO JING, CHEN LIDIAN. Electroacupuncture promotes neurological functional recovery via the retinoic acid signaling pathway in rats following cerebral ischemia-reperfusion injury. Int J Mol Med 2012; 31:225-31. [DOI: 10.3892/ijmm.2012.1166] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 09/18/2012] [Indexed: 11/05/2022] Open
|
39
|
Shearer KD, Stoney PN, Morgan PJ, McCaffery PJ. A vitamin for the brain. Trends Neurosci 2012; 35:733-41. [PMID: 22959670 DOI: 10.1016/j.tins.2012.08.005] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Revised: 08/14/2012] [Accepted: 08/14/2012] [Indexed: 11/25/2022]
Abstract
In the central nervous system (CNS) the function of retinoic acid, the active metabolite of vitamin A, is best understood from its action in guiding embryonic development; as development comes to completion, retinoic acid signaling declines. However, it is increasingly recognized that this signaling mechanism does not disappear in the adult brain but becomes more regionally focused and takes on new roles. These functions are often tied to processes of neural plasticity whether in the hippocampus, through homeostatic neural plasticity, the olfactory bulb or the hypothalamus. The role of retinoic acid in the control of plastic processes has led to suggestions of its involvement in neural disorders, both degenerative and psychiatric. This review presents a snapshot of developments in these areas over recent years.
Collapse
Affiliation(s)
- Kirsty D Shearer
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | | | | | | |
Collapse
|
40
|
Reden J, Lill K, Zahnert T, Haehner A, Hummel T. Olfactory function in patients with postinfectious and posttraumatic smell disorders before and after treatment with vitamin A: A double-blind, placebo-controlled, randomized clinical trial. Laryngoscope 2012; 122:1906-9. [DOI: 10.1002/lary.23405] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/20/2012] [Accepted: 04/11/2012] [Indexed: 11/10/2022]
|
41
|
Topographical analysis of the subependymal zone neurogenic niche. PLoS One 2012; 7:e38647. [PMID: 22745673 PMCID: PMC3379980 DOI: 10.1371/journal.pone.0038647] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 05/13/2012] [Indexed: 12/18/2022] Open
Abstract
The emerging model for the adult subependymal zone (SEZ) cell population indicates that neuronal diversity is not generated from a uniform pool of stem cells but rather from diverse and spatially confined stem cell populations. Hence, when analysing SEZ proliferation, the topography along the anterior-posterior and dorsal-ventral axes must be taken into account. However, to date, no studies have assessed SEZ proliferation according to topographical specificities and, additionally, SEZ studies in animal models of neurological/psychiatric disorders often fail to clearly specify the SEZ coordinates. This may render difficult the comparison between studies and yield contradictory results. More so, by focusing in a single spatial dimension of the SEZ, relevant findings might pass unnoticed. In this study we characterized the neural stem cell/progenitor population and its proliferation rates throughout the rat SEZ anterior-posterior and dorsal-ventral axes. We found that SEZ proliferation decreases along the anterior-posterior axis and that proliferative rates vary considerably according to the position in the dorsal-ventral axis. These were associated with relevant gradients in the neuroblasts and in the neural stem cell populations throughout the dorsal-ventral axis. In addition, we observed spatially dependent differences in BrdU/Ki67 ratios that suggest a high variability in the proliferation rate and cell cycle length throughout the SEZ; in accordance, estimation of the cell cycle length of the neuroblasts revealed shorter cell cycles at the dorsolateral SEZ. These findings highlight the need to establish standardized procedures of SEZ analysis. Herein we propose an anatomical division of the SEZ that should be considered in future studies addressing proliferation in this neural stem cell niche.
Collapse
|
42
|
Goodman T, Crandall JE, Nanescu SE, Quadro L, Shearer K, Ross A, McCaffery P. Patterning of retinoic acid signaling and cell proliferation in the hippocampus. Hippocampus 2012; 22:2171-83. [PMID: 22689466 PMCID: PMC3505796 DOI: 10.1002/hipo.22037] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2012] [Indexed: 12/23/2022]
Abstract
The nuclear receptor ligand retinoic acid (RA) has been identified as an endogenous regulatory factor in the hippocampus, acting on pyramidal neurons and granule neuron progenitors, but almost nothing is known about the distribution of RA itself in the hippocampus. This study describes the source of RA for the rodent hippocampus in the meninges via the key RA synthetic enzyme retinaldehyde dehydrogenase 2 (RALDH2). Diffusion of RA from the meninges potentially creates a gradient of RA across the infrapyramidal and suprapyramidal blades of the dentate gyrus, enhanced by the expression of the RA catabolic enzyme Cyp26B1 between the blades, and an infrapyramidal and suprapyramidal blade difference is evident in RA-regulated transcription. This asymmetry may contribute to some of the physiological and molecular differences between the blades, including a disparity in the rates of cell proliferation in the subgranular zone of the two blades through RA inhibition of cell proliferation. Such differences can be altered by either the application of excess RA, its effect dependent on the relative position along the septotemporal axis, or change in RA signaling through mutation of retinol binding protein, while the capacity of RA to inhibit proliferation of cells in the dentate gyrus is demonstrated using in vitro slice culture. Use of synthetic and catabolic enzymes in the hippocampus to create differing zones of RA concentration parallels the mechanisms used in the developing brain to generate patterns of RA-regulated transcription. © 2012 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Timothy Goodman
- Institute of Medical Sciences, University of Aberdeen, Aberdeenshire, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Retinoic acid (RA) is a vitamin A-derived, non-peptidic, small lipophilic molecule that acts as ligand for nuclear RA receptors (RARs), converting them from transcriptional repressors to activators. The distribution and levels of RA in embryonic tissues are tightly controlled by regulated synthesis through the action of specific retinol and retinaldehyde dehydrogenases and by degradation via specific cytochrome P450s (CYP26s). Recent studies indicate that RA action involves an interplay between diffusion (morphogen-like) gradients and the establishment of signalling boundaries due to RA metabolism, thereby allowing RA to finely control the differentiation and patterning of various stem/progenitor cell populations. Here, we provide an overview of the RA biosynthesis, degradation and signalling pathways and review the main functions of this molecule during embryogenesis.
Collapse
Affiliation(s)
- Muriel Rhinn
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.
| | | |
Collapse
|
44
|
Chan TJ, Her LS, Liaw HJ, Chen MC, Tzeng SF. Retinoic acid mediates the expression of glutamate transporter-1 in rat astrocytes through genomic RXR action and non-genomic protein kinase C signaling pathway. J Neurochem 2012; 121:537-50. [PMID: 22380620 DOI: 10.1111/j.1471-4159.2012.07715.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Astrocytic glutamate transporter-1 (GLT-1) is responsible for 90% of forebrain glutamate uptake in the adult CNS. Retinoic acid (RA) is a potent regulator of neural cell differentiation and neuronal maturation in the developing CNS through activation of RA receptors/retinoic X receptors (RXRs) or non-genomic mechanisms. Although rat GLT-1 contains several RXR binding regions, RA-triggered RXR mechanisms regulating GLT-1 expression remain unknown. RA applied at submicromolar concentrations for 24 h significantly reduced GLT-1 mRNA and membrane levels in astrocytes and dibutyryl cAMP (dbcAMP)-primed astrocytes. An RXR agonist reduced astrocytic GLT-1 mRNA expression, whereas an RXR antagonist blocked the effects of RA on the reduction of astrocytic GLT-1 mRNA expression. Electrophoresis motility shift assay indicated that RA-treatment increased astrocytic RXR-DNA binding activity. RA-induced reduction in GLT-1 mRNA expression was also observed in dbcAMP-primed astrocytes. Through lentivirus-mediated astrocytic over-expression of rat GLT-1, levels of GLT-1 in the processes of dbcAMP-treated astrocytes were attenuated by exposure to RA. The protein kinase C inhibitor, Bis I, restored GLT-1 distribution in the processes of RA-treated dbcAMP-primed astrocytes. These results suggest that RA reduces astrocytic GLT-1 levels through both RXR-mediated inhibition at the transcriptional level and triggering activation of protein kinase C which reduces cell surface GLT-1 levels.
Collapse
Affiliation(s)
- Ti-Ju Chan
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
45
|
Zappaterra MW, Lehtinen MK. The cerebrospinal fluid: regulator of neurogenesis, behavior, and beyond. Cell Mol Life Sci 2012; 69:2863-78. [PMID: 22415326 DOI: 10.1007/s00018-012-0957-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 12/11/2022]
Abstract
The cerebrospinal fluid (CSF) has attracted renewed interest as an active signaling milieu that regulates brain development, homeostasis, and disease. Advances in proteomics research have enabled an improved characterization of the CSF from development through adulthood, and key neurogenic signaling pathways that are transmitted via the CSF are now being elucidated. Due to its immediate contact with neural stem cells in the developing and adult brain, the CSF's ability to swiftly distribute signals across vast distances in the central nervous system is opening avenues to novel and exciting therapeutic approaches. In this review, we will discuss the development of the choroid plexus-CSF system, and review the current literature on how the CSF actively regulates mammalian brain development, behavior, and responses to traumatic brain injury.
Collapse
Affiliation(s)
- Mauro W Zappaterra
- Department of Physical Medicine and Rehabilitation, VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, Los Angeles, CA 90073, USA.
| | | |
Collapse
|
46
|
Paschaki M, Lin SC, Wong RLY, Finnell RH, Dollé P, Niederreither K. Retinoic acid-dependent signaling pathways and lineage events in the developing mouse spinal cord. PLoS One 2012; 7:e32447. [PMID: 22396766 PMCID: PMC3292566 DOI: 10.1371/journal.pone.0032447] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 01/26/2012] [Indexed: 11/19/2022] Open
Abstract
Studies in avian models have demonstrated an involvement of retinoid signaling in early neural tube patterning. The roles of this signaling pathway at later stages of spinal cord development are only partly characterized. Here we use Raldh2-null mouse mutants rescued from early embryonic lethality to study the consequences of lack of endogenous retinoic acid (RA) in the differentiating spinal cord. Mid-gestation RA deficiency produces prominent structural and molecular deficiencies in dorsal regions of the spinal cord. While targets of Wnt signaling in the dorsal neuronal lineage are unaltered, reductions in Fibroblast Growth Factor (FGF) and Notch signaling are clearly observed. We further provide evidence that endogenous RA is capable of driving stem cell differentiation. Raldh2 deficiency results in a decreased number of spinal cord derived neurospheres, which exhibit a reduced differentiation potential. Raldh2-null neurospheres have a decreased number of cells expressing the neuronal marker β-III-tubulin, while the nestin-positive cell population is increased. Hence, in vivo retinoid deficiency impaired neural stem cell growth. We propose that RA has separable functions in the developing spinal cord to (i) maintain high levels of FGF and Notch signaling and (ii) drive stem cell differentiation, thus restricting both the numbers and the pluripotent character of neural stem cells.
Collapse
Affiliation(s)
- Marie Paschaki
- Development and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Song-Chang Lin
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rebecca Lee Yean Wong
- Center for Environmental and Genetic Medicine, Institute of Biosciences and Technology, The Texas A&M University System Health Science Center, Houston, Texas, United States of America
| | - Richard H. Finnell
- Department of Nutritional Sciences, Dell Pediatric Research Institute, University of Texas, Austin, Texas, United States of America
| | - Pascal Dollé
- Development and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Karen Niederreither
- Department of Nutritional Sciences, Dell Pediatric Research Institute, University of Texas, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
47
|
Molecular regulation of striatal development: a review. ANATOMY RESEARCH INTERNATIONAL 2012; 2012:106529. [PMID: 22567304 PMCID: PMC3335634 DOI: 10.1155/2012/106529] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 10/07/2011] [Indexed: 01/26/2023]
Abstract
The central nervous system is composed of the brain and the spinal cord. The brain is a complex organ that processes and coordinates activities of the body in bilaterian, higher-order animals. The development of the brain mirrors its complex function as it requires intricate genetic signalling at specific times, and deviations from this can lead to brain malformations such as anencephaly. Research into how the CNS is specified and patterned has been studied extensively in chick, fish, frog, and mice, but findings from the latter will be emphasised here as higher-order mammals show most similarity to the human brain. Specifically, we will focus on the embryonic development of an important forebrain structure, the striatum (also known as the dorsal striatum or neostriatum). Over the past decade, research on striatal development in mice has led to an influx of new information about the genes involved, but the precise orchestration between the genes, signalling molecules, and transcription factors remains unanswered. We aim to summarise what is known to date about the tightly controlled network of interacting genes that control striatal development. This paper will discuss early telencephalon patterning and dorsal ventral patterning with specific reference to the genes involved in striatal development.
Collapse
|
48
|
Dranse HJ, Sampaio AV, Petkovich M, Underhill TM. Genetic deletion of Cyp26b1 negatively impacts limb skeletogenesis by inhibiting chondrogenesis. J Cell Sci 2011; 124:2723-34. [PMID: 21807937 DOI: 10.1242/jcs.084699] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Cyp26b1, a retinoic acid (RA)-metabolising enzyme, is expressed in the developing limb bud, and Cyp26b1(-/-) mice present with severe limb defects. These malformations might be attributable to an RA-induced patterning defect; however, recent reports suggest that RA is dispensable for limb patterning. In this study, we examined the role of endogenous retinoid signalling in skeletogenesis using Cyp26b1(-/-) mice and transgenic mice in which Cyp26b1 is conditionally deleted under control of the Prrx1 promoter beginning at ~E9.5 (Prrx1Cre(+)/Cyp26b1(fl/fl)). We found that the limb phenotype in Prrx1Cre(+)/Cyp26b1(fl/fl) mice was less severe than that observed in Cyp26b1(-/-) animals and that a change in retinoid signalling contributed to the difference in phenotypes. We systematically examined the role of endogenous RA signalling in chondrogenesis and found that Cyp26b1(-/-) cells and limb mesenchymal cells treated with a CYP inhibitor, are maintained in a pre-chondrogenic state, exhibit reduced chondroblast differentiation and have modestly accelerated chondrocyte hypertrophy. Furthermore, Cyp26b1(-/-) mesenchyme exhibited an increase in expression of genes in a closely related tendogenic lineage, indicating that retinoid signals in the limb interfere with differentiation and maintain progenitor status. Together, these findings support an important function for RA in regulating the behaviour of mesenchymal progenitors, and their subsequent differentiation and maturation.
Collapse
Affiliation(s)
- Helen J Dranse
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | | | | |
Collapse
|
49
|
Paulsen BS, Souza CS, Chicaybam L, Bonamino MH, Bahia M, Costa SL, Borges HL, Rehen SK. Agathisflavone Enhances Retinoic Acid-Induced Neurogenesis and Its Receptors α and β in Pluripotent Stem Cells. Stem Cells Dev 2011; 20:1711-21. [DOI: 10.1089/scd.2010.0446] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Bruna S. Paulsen
- Instituto de Ciências Biomédicas, Laboratório Nacional de Células-Tronco Embrionárias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleide S. Souza
- Instituto de Ciências Biomédicas, Laboratório Nacional de Células-Tronco Embrionárias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Leonardo Chicaybam
- Divisão de Medicina Experimental, Instituto Nacional do Câncer, Rio de Janeiro, Brazil
| | | | - Marcus Bahia
- Universidade Federal do Recôncavo da Bahia, Recôncavo, Brazil
| | - Silvia Lima Costa
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Helena L. Borges
- Instituto de Ciências Biomédicas, Laboratório Nacional de Células-Tronco Embrionárias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stevens K. Rehen
- Instituto de Ciências Biomédicas, Laboratório Nacional de Células-Tronco Embrionárias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
50
|
Abstract
Cerebral cortical progenitor cells can be classified into several different types, and each progenitor type integrates cell-intrinsic and cell-extrinsic cues to regulate neurogenesis. On one hand, cell-intrinsic mechanisms that depend upon appropriate apical-basal polarity are established by adherens junctions and apical complex proteins and are particularly important in progenitors with apical processes contacting the lateral ventricle. The apical protein complexes themselves are concentrated at the ventricular surface, and apical complex proteins regulate mitotic spindle orientation and cell fate. On the other hand, remarkably little is known about how cell-extrinsic cues signal to progenitors and couple with cell-intrinsic mechanisms to instruct neurogenesis. Recent research shows that the cerebrospinal fluid, which contacts apical progenitors at the ventricular surface and bathes the apical complex of these cells, provides growth- and survival-promoting cues for neural progenitor cells in developing and adult brain. This review addresses how the apical-basal polarity of progenitor cells regulates cell fate and allows progenitors to sample diffusible signals distributed by the cerebrospinal fluid. We also review several classes of signaling factors that the cerebrospinal fluid distributes to the developing brain to instruct neurogenesis.
Collapse
Affiliation(s)
- Maria K Lehtinen
- Division of Genetics, Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|