51
|
Glial cell type-specific gene expression in the mouse cerebrum using the piggyBac system and in utero electroporation. Sci Rep 2021; 11:4864. [PMID: 33649472 PMCID: PMC7921133 DOI: 10.1038/s41598-021-84210-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glial cells such as astrocytes and oligodendrocytes play crucial roles in the central nervous system. To investigate the molecular mechanisms underlying the development and the biological functions of glial cells, simple and rapid techniques for glial cell-specific genetic manipulation in the mouse cerebrum would be valuable. Here we uncovered that the Gfa2 promoter is suitable for selective gene expression in astrocytes when used with the piggyBac system and in utero electroporation. In contrast, the Blbp promoter, which has been used to induce astrocyte-specific gene expression in transgenic mice, did not result in astrocyte-specific gene expression. We also identified the Plp1 and Mbp promoters could be used with the piggyBac system and in utero electroporation to induce selective gene expression in oligodendrocytes. Furthermore, using our technique, neuron-astrocyte or neuron-oligodendrocyte interactions can be visualized by labeling neurons, astrocytes and oligodendrocytes differentially. Our study provides a fundamental basis for specific transgene expression in astrocytes and/or oligodendrocytes in the mouse cerebrum.
Collapse
|
52
|
|
53
|
Franchini LF. Genetic Mechanisms Underlying Cortical Evolution in Mammals. Front Cell Dev Biol 2021; 9:591017. [PMID: 33659245 PMCID: PMC7917222 DOI: 10.3389/fcell.2021.591017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The remarkable sensory, motor, and cognitive abilities of mammals mainly depend on the neocortex. Thus, the emergence of the six-layered neocortex in reptilian ancestors of mammals constitutes a fundamental evolutionary landmark. The mammalian cortex is a columnar epithelium of densely packed cells organized in layers where neurons are generated mainly in the subventricular zone in successive waves throughout development. Newborn cells move away from their site of neurogenesis through radial or tangential migration to reach their specific destination closer to the pial surface of the same or different cortical area. Interestingly, the genetic programs underlying neocortical development diversified in different mammalian lineages. In this work, I will review several recent studies that characterized how distinct transcriptional programs relate to the development and functional organization of the neocortex across diverse mammalian lineages. In some primates such as the anthropoids, the neocortex became extremely large, especially in humans where it comprises around 80% of the brain. It has been hypothesized that the massive expansion of the cortical surface and elaboration of its connections in the human lineage, has enabled our unique cognitive capacities including abstract thinking, long-term planning, verbal language and elaborated tool making capabilities. I will also analyze the lineage-specific genetic changes that could have led to the modification of key neurodevelopmental events, including regulation of cell number, neuronal migration, and differentiation into specific phenotypes, in order to shed light on the evolutionary mechanisms underlying the diversity of mammalian brains including the human brain.
Collapse
Affiliation(s)
- Lucía Florencia Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
54
|
NMDA Receptor Enhances Correlation of Spontaneous Activity in Neonatal Barrel Cortex. J Neurosci 2021; 41:1207-1217. [PMID: 33372060 DOI: 10.1523/jneurosci.0527-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 01/07/2023] Open
Abstract
Correlated spontaneous activity plays critical role in the organization of neocortical circuits during development. However, cortical mechanisms regulating activity correlation are still elusive. In this study, using two-photon calcium imaging of the barrel cortex layer 4 (L4) in living neonatal mice, we found that NMDA receptors (NMDARs) in L4 neurons are important for enhancement of spontaneous activity correlation. Disruption of GluN1 (Grin1), an obligatory NMDAR subunit, in a sparse population of L4 neurons reduced activity correlation between GluN1 knock-out (GluN1KO) neuron pairs within a barrel. This reduction in activity correlation was even detected in L4 neuron pairs in neighboring barrels and most evident when either or both of neurons are located on the barrel edge. Our results provide evidence for the involvement of L4 neuron NMDARs in spatial organization of the spontaneous firing activity of L4 neurons in the neonatal barrel cortex.SIGNIFICANCE STATEMENT Precise wiring of the thalamocortical circuits is necessary for proper sensory information processing, and thalamus-derived correlated spontaneous activity is important for thalamocortical circuit formation. The molecular mechanisms involved in the correlated activity transfer from the thalamus to the neocortex are largely unknown. In vivo two-photon calcium imaging of the neonatal barrel cortex revealed that correlated spontaneous activity between layer four neurons is reduced by mosaic knock-out (KO) of the NMDA receptor (NMDAR) obligatory subunit GluN1. Our results suggest that the function of NMDARs in layer four neurons is necessary for the communication between presynaptic and postsynaptic partners during thalamocortical circuit formation.
Collapse
|
55
|
Kumar S, Voracek M, Singh M. The effects of hand preference and sex on right-left asymmetry in dorsal digit lengths among adults and children. Early Hum Dev 2021; 153:105293. [PMID: 33340946 DOI: 10.1016/j.earlhumdev.2020.105293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Right-hand preference is related to stronger right-directional asymmetry in the length of proximal upper-limb bones, although the relationships of hand preference with directional asymmetry in phalangeal bone lengths are not known. Furthermore, dorsal digit length is an easy-to-measure, faithful proxy of X-rayed phalangeal bone length (which is costly and difficult to measure). AIM To study the effects of hand preference, sex, and age on right-left (R-L) asymmetry in dorsal digit lengths. METHODS We measured all dorsal digit lengths (except the thumb) in comparable numbers of left-handers and right-handers in samples of adults (N = 151, age: M = 22.6 years, SD = 3.3) and children (N = 65, age: M = 5.0 years, SD = 1.0). RESULTS Right-handers and adults had stronger right-directional asymmetry in digit lengths than left-handers and children. A Bayesian analysis yielded an 'extremely strong likelihood' of no sex differences in the R-L asymmetry of dorsal digit lengths 2 and 4. CONCLUSIONS The effects of hand preference, sex, and age on R-L asymmetry appear to be similar for phalangeal bone length and other (proximal) upper-limb bone lengths. Two distinct biologic mechanisms (i.e., a general right-directional asymmetry mechanism and a handedness-related directional asymmetry mechanism) may contribute to observed R-L asymmetry in limbs. Fingertip fat and bone digit length do not seem to contribute to sex differences in the R-L asymmetry (Dr-l) of the widely studied second-to-fourth digit ratio (2D:4D).
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Psychology, D.A.V. College, Muzaffarnagar, Uttar Pradesh, India.
| | - Martin Voracek
- Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, Austria.
| | - Maharaj Singh
- Department of Research and Graduate Studies, School of Dentistry, Marquette University, Milwaukee, WI, USA.
| |
Collapse
|
56
|
Sensational developments in somatosensory development? Curr Opin Neurobiol 2021; 66:212-223. [PMID: 33454646 DOI: 10.1016/j.conb.2020.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/25/2022]
Abstract
This is an overview of the most recent advances pertaining to the development of the cardinal components of the somatosensory system: the peripheral sensory neurons that perceive somatosensory stimuli, the first line central nervous system circuits that modulate them, and the higher structures such as the somatosensory cortex that eventually compute a motor response to them. Here, I also review the most recent findings concerning the role of neuronal activity in somatosensory development, formation of somatotopic maps, insights into human somatosensory development and the link between aberrant somatosensation and neurodevelopmental disorders.
Collapse
|
57
|
Tsunekawa Y, Terhune RK, Matsuzaki F. Protocol for De Novo Gene Targeting Via In Utero Electroporation. Methods Mol Biol 2021; 2312:309-320. [PMID: 34228299 DOI: 10.1007/978-1-0716-1441-9_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Developments in genome-editing technology, especially CRISPR-Cas9, have revolutionized the way in which genetically engineered animals are generated. However, the process of generation includes microinjection to the one-cell stage embryo and the transfer of the microinjected embryo to the surrogate animals, which requires trained personnel. We recently reported the method includes introduction of CRISPR-Cas9 systems to the developing cerebral cortex via in utero electroporation thus generating gene-targeted neural stem cells in vivo. This technique is widely applicable for gene knockout, monitoring gene expression, and lineage analysis in developmental biology. In this chapter, the detailed protocol of EGFP (enhanced green fluorescent protein) knock-in method via in utero electroporation is described.
Collapse
Affiliation(s)
- Yuji Tsunekawa
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
- Division of Molecular and Medical Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| | | | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
- Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
58
|
Mollinedo-Gajate I, Song C, Knöpfel T. Genetically Encoded Voltage Indicators. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:209-224. [PMID: 33398815 DOI: 10.1007/978-981-15-8763-4_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Optogenetic approaches combine the power to allocate optogenetic tools (proteins) to specific cell populations (defined genetically or functionally) and the use of light-based interfaces between biological wetware (cells and tissues) and hardware (controllers and recorders). The optogenetic toolbox contains two main compartments: tools to interfere with cellular processes and tools to monitor cellular events. Among the latter are genetically encoded voltage indicators (GEVIs). This chapter outlines the development, current state of the art and prospects of emerging optical GEVI imaging technologies.
Collapse
Affiliation(s)
| | - Chenchen Song
- Laboratory for Neuronal Circuit Dynamics, Imperial College London, London, UK
| | - Thomas Knöpfel
- Laboratory for Neuronal Circuit Dynamics, Imperial College London, London, UK.
| |
Collapse
|
59
|
Rao MS, Mizuno H. Elucidating mechanisms of neuronal circuit formation in layer 4 of the somatosensory cortex via intravital imaging. Neurosci Res 2020; 167:47-53. [PMID: 33309867 DOI: 10.1016/j.neures.2020.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
The cerebral cortex has complex yet perfectly wired neuronal circuits that are important for high-level brain functions such as perception and cognition. The rodent's somatosensory system is widely used for understanding the mechanisms of circuit formation during early developmental periods. In this review, we summarize the developmental processes of circuit formation in layer 4 of the somatosensory cortex, and we describe the molecules involved in layer 4 circuit formation and neuronal activity-dependent mechanisms of circuit formation. We also introduce the dynamic mechanisms of circuit formation in layer 4 revealed by intravital two-photon imaging technologies, which include time-lapse imaging of neuronal morphology and calcium imaging of neuronal activity in newborn mice.
Collapse
Affiliation(s)
- Madhura S Rao
- Laboratory of Multi-dimensional Imaging, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, 860-0811, Japan; Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Hidenobu Mizuno
- Laboratory of Multi-dimensional Imaging, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, 860-0811, Japan; Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.
| |
Collapse
|
60
|
Effects of hand preference on digit lengths and digit ratios among children and adults. Early Hum Dev 2020; 151:105204. [PMID: 33059164 DOI: 10.1016/j.earlhumdev.2020.105204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 01/21/2023]
Abstract
BACKGROUND Prenatal sex hormones may not exclusively determine effects of hand preference on digit ratios. Genetic determination is an alternative possibility. AIM To study the likelihood of similar effects of hand preference on digit lengths and digit ratios. METHODS We selected similar numbers of left-handers and right-handers in samples of kindergarten children (N = 101, age range: 3.5-7 years) and adults (N = 189, age range: 17-28 years) and measured digit lengths (excluding the thumb) directly on the palmar hand. RESULTS Compared to right-handers, left-handers had longer digits and lower third-to-fourth (3D:4D) digit ratios among children, whereas an opposite pattern of handedness differences occurred among adults. CONCLUSIONS Effects of hand preference on digit lengths and ratios might be genetically/ontogenetically determined. Also discussed are implications of this set of findings for digit ratio research.
Collapse
|
61
|
Diaz C, Puelles L. Developmental Genes and Malformations in the Hypothalamus. Front Neuroanat 2020; 14:607111. [PMID: 33324176 PMCID: PMC7726113 DOI: 10.3389/fnana.2020.607111] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus is a heterogeneous rostral forebrain region that regulates physiological processes essential for survival, energy metabolism, and reproduction, mainly mediated by the pituitary gland. In the updated prosomeric model, the hypothalamus represents the rostralmost forebrain, composed of two segmental regions (terminal and peduncular hypothalamus), which extend respectively into the non-evaginated preoptic telencephalon and the evaginated pallio-subpallial telencephalon. Complex genetic cascades of transcription factors and signaling molecules rule their development. Alterations of some of these molecular mechanisms acting during forebrain development are associated with more or less severe hypothalamic and pituitary dysfunctions, which may be associated with brain malformations such as holoprosencephaly or septo-optic dysplasia. Studies on transgenic mice with mutated genes encoding critical transcription factors implicated in hypothalamic-pituitary development are contributing to understanding the high clinical complexity of these pathologies. In this review article, we will analyze first the complex molecular genoarchitecture of the hypothalamus resulting from the activity of previous morphogenetic signaling centers and secondly some malformations related to alterations in genes implicated in the development of the hypothalamus.
Collapse
Affiliation(s)
- Carmen Diaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| |
Collapse
|
62
|
Ratzan EM, Moon AM, Deans MR. Fgf8 genetic labeling reveals the early specification of vestibular hair cell type in mouse utricle. Development 2020; 147:dev.192849. [PMID: 33046506 DOI: 10.1242/dev.192849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/07/2020] [Indexed: 01/16/2023]
Abstract
FGF8 signaling plays diverse roles in inner ear development, acting at multiple stages from otic placode induction to cellular differentiation in the organ of Corti. As a secreted morphogen with diverse functions, Fgf8 expression is likely to be spatially restricted and temporally dynamic throughout inner ear development. We evaluated these characteristics using genetic labeling mediated by Fgf8 mcm gene-targeted mice and determined that Fgf8 expression is a specific and early marker of Type-I vestibular hair cell identity. Fgf8 mcm expression initiates at E11.5 in the future striolar region of the utricle, labeling hair cells following EdU birthdating, and demonstrates that sub-type identity is determined shortly after terminal mitosis. This early fate specification is not apparent using markers or morphological criteria that are not present before birth in the mouse. Although analyses of Fgf8 conditional knockout mice did not reveal developmental phenotypes, the restricted pattern of Fgf8 expression suggests that functionally redundant FGF ligands may contribute to vestibular hair cell differentiation and supports a developmental model in which Type-I and Type-II hair cells develop in parallel rather than from an intermediate precursor.
Collapse
Affiliation(s)
- Evan M Ratzan
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.,Interdepartmental Program in Neuroscience, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Anne M Moon
- Departments of Molecular and Functional Genomics and Pediatrics, Weis Center for Research, Geisinger Clinic and Geisinger Commonwealth School of Medicine, Danville, PA 17822, USA.,Departments of Pediatrics and Human Genetics, University of Utah, Salt Lake City, UT 84112 USA
| | - Michael R Deans
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA .,Department of Surgery, Division of Otolaryngology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
63
|
Chiaradia I, Lancaster MA. Brain organoids for the study of human neurobiology at the interface of in vitro and in vivo. Nat Neurosci 2020; 23:1496-1508. [PMID: 33139941 DOI: 10.1038/s41593-020-00730-3] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
Brain development is an extraordinarily complex process achieved through the spatially and temporally regulated release of key patterning factors. In vitro neurodevelopmental models seek to mimic these processes to recapitulate the steps of tissue fate acquisition and morphogenesis. Classic two-dimensional neural cultures present higher homogeneity but lower complexity compared to the brain. Brain organoids instead have more advanced cell composition, maturation and tissue architecture. They can thus be considered at the interface of in vitro and in vivo neurobiology, and further improvements in organoid techniques are continuing to narrow the gap with in vivo brain development. Here we describe these efforts to recapitulate brain development in neural organoids and focus on their applicability for disease modeling, evolutionary studies and neural network research.
Collapse
Affiliation(s)
- Ilaria Chiaradia
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
64
|
Huang JY, Krebs BB, Miskus ML, Russell ML, Duffy EP, Graf JM, Lu HC. Enhanced FGFR3 activity in postmitotic principal neurons during brain development results in cortical dysplasia and axonal tract abnormality. Sci Rep 2020; 10:18508. [PMID: 33116259 PMCID: PMC7595096 DOI: 10.1038/s41598-020-75537-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Abnormal levels of fibroblast growth factors (FGFs) and FGF receptors (FGFRs) have been detected in various neurological disorders. The potent impact of FGF-FGFR in multiple embryonic developmental processes makes it challenging to elucidate their roles in postmitotic neurons. Taking an alternative approach to examine the impact of aberrant FGFR function on glutamatergic neurons, we generated a FGFR gain-of-function (GOF) transgenic mouse, which expresses constitutively activated FGFR3 (FGFR3K650E) in postmitotic glutamatergic neurons. We found that GOF disrupts mitosis of radial-glia neural progenitors (RGCs), inside-out radial migration of post-mitotic glutamatergic neurons, and axonal tract projections. In particular, late-born CUX1-positive neurons are widely dispersed throughout the GOF cortex. Such a cortical migration deficit is likely caused, at least in part, by a significant reduction of the radial processes projecting from RGCs. RNA-sequencing analysis of the GOF embryonic cortex reveals significant alterations in several pathways involved in cell cycle regulation and axonal pathfinding. Collectively, our data suggest that FGFR3 GOF in postmitotic neurons not only alters axonal growth of postmitotic neurons but also impairs RGC neurogenesis and radial glia processes.
Collapse
Affiliation(s)
- Jui-Yen Huang
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| | - Bruna Baumgarten Krebs
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN, 47405, USA
| | - Marisha Lynn Miskus
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - May Lin Russell
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Eamonn Patrick Duffy
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Jason Michael Graf
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
65
|
Iwasato T. In vivo imaging of neural circuit formation in the neonatal mouse barrel cortex. Dev Growth Differ 2020; 62:476-486. [DOI: 10.1111/dgd.12693] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/13/2020] [Accepted: 09/27/2020] [Indexed: 01/21/2023]
Affiliation(s)
- Takuji Iwasato
- Laboratory of Mammalian Neural Circuits National Institute of Genetics Mishima Japan
- Department of Genetics SOKENDAI Mishima Japan
| |
Collapse
|
66
|
Bartkowska K, Tepper B, Gawda A, Jarosik M, Sobolewska P, Turlejski K, Djavadian RL. Inhibition of TrkB- and TrkC-Signaling Pathways Affects Neurogenesis in the Opossum Developing Neocortex. Cereb Cortex 2020; 29:3666-3675. [PMID: 30272136 DOI: 10.1093/cercor/bhy246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022] Open
Abstract
We have previously reported that the blockage of TrkB and TrkC signaling in primary culture of opossum neocortical cells affects neurogenesis that involves a range of processes including cell proliferation, differentiation, and survival. Here, we studied whether TrkB and TrkC activity specifically affects various types of progenitor cell populations during neocortex formation in the Monodelphis opossum in vivo. We found that the inhibition of TrkB and TrkC activities affects the same proliferative cellular phenotype, but TrkC causes more pronounced changes in the rate of cell divisions. Additionally, inhibition of TrkB and TrkC does not affect apoptosis in vivo, which was found in cell culture experiments. The lack of TrkB and TrkC receptor activity caused the arrest of newly generated neurons; therefore, they could not penetrate the subplate zone. We suggest that at this time point in development, migration consists of 2 steps. During the initial step, neurons migrate and reach the base of the subplate, whereas during the next step the migration of neurons to their final position is regulated by TrkB or TrkC signaling.
Collapse
Affiliation(s)
- K Bartkowska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - B Tepper
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - A Gawda
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - M Jarosik
- Faculty of Biology and Environmental Sciences, Cardinal Stefan Wyszynski University in Warsaw, Poland
| | - P Sobolewska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - K Turlejski
- Faculty of Biology and Environmental Sciences, Cardinal Stefan Wyszynski University in Warsaw, Poland
| | - R L Djavadian
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
67
|
Linnerbauer M, Rothhammer V. Protective Functions of Reactive Astrocytes Following Central Nervous System Insult. Front Immunol 2020; 11:573256. [PMID: 33117368 PMCID: PMC7561408 DOI: 10.3389/fimmu.2020.573256] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022] Open
Abstract
Astrocytes play important roles in numerous central nervous system disorders including autoimmune inflammatory, hypoxic, and degenerative diseases such as Multiple Sclerosis, ischemic stroke, and Alzheimer’s disease. Depending on the spatial and temporal context, activated astrocytes may contribute to the pathogenesis, progression, and recovery of disease. Recent progress in the dissection of transcriptional responses to varying forms of central nervous system insult has shed light on the mechanisms that govern the complexity of reactive astrocyte functions. While a large body of research focuses on the pathogenic effects of reactive astrocytes, little is known about how they limit inflammation and contribute to tissue regeneration. However, these protective astrocyte pathways might be of relevance for the understanding of the underlying pathology in disease and may lead to novel targeted approaches to treat autoimmune inflammatory and degenerative disorders of the central nervous system. In this review article, we have revisited the emerging concept of protective astrocyte functions and discuss their role in the recovery from inflammatory and ischemic disease as well as their role in degenerative disorders. Focusing on soluble astrocyte derived mediators, we aggregate the existing knowledge on astrocyte functions in the maintenance of homeostasis as well as their reparative and tissue-protective function after acute lesions and in neurodegenerative disorders. Finally, we give an outlook of how these mediators may guide future therapeutic strategies to tackle yet untreatable disorders of the central nervous system.
Collapse
Affiliation(s)
- Mathias Linnerbauer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Veit Rothhammer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
68
|
Erzurumlu RS, Gaspar P. How the Barrel Cortex Became a Working Model for Developmental Plasticity: A Historical Perspective. J Neurosci 2020; 40:6460-6473. [PMID: 32817388 PMCID: PMC7486654 DOI: 10.1523/jneurosci.0582-20.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 01/08/2023] Open
Abstract
For half a century now, the barrel cortex of common laboratory rodents has been an exceptionally useful model for studying the formation of topographically organized maps, neural patterning, and plasticity, both in development and in maturity. We present a historical perspective on how barrels were discovered, and how thereafter, they became a workhorse for developmental neuroscientists and for studies on brain plasticity and activity-dependent modeling of brain circuits. What is particularly remarkable about this sensory system is a cellular patterning that is induced by signals derived from the sensory receptors surrounding the snout whiskers and transmitted centrally to the brainstem (barrelettes), the thalamus (barreloids), and the neocortex (barrels). Injury to the sensory receptors shortly after birth leads to predictable pattern alterations at all levels of the system. Mouse genetics have increased our understanding of how barrels are constructed and revealed the interplay of the molecular programs that direct axon growth and cell specification, with activity-dependent mechanisms. There is an ever-rising interest in this sensory system as a neurobiological model to study development of somatotopy, patterning, and plasticity at both the morphologic and physiological levels. This article is part of a group of articles commemorating the 50th anniversary of the Society for Neuroscience.
Collapse
Affiliation(s)
- Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Patricia Gaspar
- Institut National de la Santé et de la Recherche Médicale, Paris Brain Institute, Sorbonne Universités, Paris, France 75013
| |
Collapse
|
69
|
Schohl A, Chorghay Z, Ruthazer ES. A Simple and Efficient Method for Visualizing Individual Cells in vivo by Cre-Mediated Single-Cell Labeling by Electroporation (CREMSCLE). Front Neural Circuits 2020; 14:47. [PMID: 32848634 PMCID: PMC7399061 DOI: 10.3389/fncir.2020.00047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/08/2020] [Indexed: 11/13/2022] Open
Abstract
Efficient methods for visualizing cell morphology in the intact animal are of great benefit to the study of structural development in the nervous system. Quantitative analysis of the complex arborization patterns of brain cells informs cell-type classification, dissection of neuronal circuit wiring, and the elucidation of growth and plasticity mechanisms. Time-lapse single-cell morphological analysis requires labeling and imaging of single cells in situ without contamination from the ramified processes of other nearby cells. Here, using the Xenopus laevis optic tectum as a model system, we describe CRE-Mediated Single-Cell Labeling by Electroporation (CREMSCLE), a technique we developed based on bulk co-electroporation of Cre-dependent inducible expression vectors, together with very low concentrations of plasmid encoding Cre recombinase. This method offers efficient, sparse labeling in any brain area where bulk electroporation is possible. Unlike juxtacellular single-cell electroporation methods, CREMSCLE relies exclusively on the bulk electroporation technique, circumventing the need to precisely position a micropipette next to the target cell. Compared with viral transduction methods, it is fast and safe, generating high levels of expression within 24 h of introducing non-infectious plasmid DNA. In addition to increased efficiency of single-cell labeling, we confirm that CREMSCLE also allows for efficient co-expression of multiple gene products in the same cell. Furthermore, we demonstrate that this method is particularly well-suited for labeling immature neurons to follow their maturation over time. This approach therefore lends itself well to time-lapse morphological studies, particularly in the context of early neuronal development and under conditions that prevent more difficult visualized juxtacellular electroporation.
Collapse
Affiliation(s)
- Anne Schohl
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Zahraa Chorghay
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Edward S Ruthazer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
70
|
Pattabiraman K, Muchnik SK, Sestan N. The evolution of the human brain and disease susceptibility. Curr Opin Genet Dev 2020; 65:91-97. [PMID: 32629339 DOI: 10.1016/j.gde.2020.05.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/05/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022]
Abstract
Evolutionary perspective is critical for understanding human biology, human medicine, and the traits that make human beings unique. One of the crucial characteristics that sets humans apart from other extant species is our cognitive ability, which allows for complex processes including symbolic thought, theory of mind, and syntactical-grammatical language, and is thought to arise from the expansion and specialization of the human nervous system. It has been hypothesized that the same evolutionary changes that allowed us to develop these valuable skills made humans susceptible to neurodevelopmental and neurodegenerative disease. Unfortunately, our lack of access to our extinct ancestors makes this a difficult hypothesis to test, but recent collaborations between the fields of evolution, genetics, genomics, neuroscience, neurology and psychiatry have begun to provide some clues. Here, we will outline recent work in those fields that have utilized our growing knowledge of disease risk genes and loci, identified by wide-scale genetic studies, and nervous system development and function to draw conclusions about the impact of human-specific aspects of evolution. We will discuss studies that assess evolution at a variety of scales including at the levels of whole brain regions, cell types, synapses, metabolic processes, gene expression patterns, and gene regulation. At all of these levels, there is preliminary evidence that human-specific brain features are linked to neurodevelopmental and neurodegenerative disease risk.
Collapse
Affiliation(s)
- Kartik Pattabiraman
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Yale Child Study Center, New Haven, CT 06510, USA
| | - Sydney Keaton Muchnik
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Yale Child Study Center, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Psychiatry and Comparative Medicine, Kavli Institute for Neuroscience, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
71
|
Abstract
The cerebral cortex is composed of an exquisitely complex network of interconnected neurons supporting the higher cognitive functions of the brain. Here, we provide a fully detailed, step-by-step protocol to perform in utero cortical electroporation of plasmids, a simple surgical procedure designed to manipulate gene expression in a subset of glutamatergic pyramidal cortical neurons in vivo. This method has been used to visualize defects in neuronal migration, axon projections, terminal axon branching, or dendrite and synapse development. For complete details on the use and execution of this protocol, please refer to Courchet et al., 2013, Mairet-Coello et al., 2013 or Shimojo et al. (2015).
Collapse
Affiliation(s)
- Géraldine Meyer-Dilhet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| | - Julien Courchet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| |
Collapse
|
72
|
Fedorchak NJ, Iyer N, Ashton RS. Bioengineering tissue morphogenesis and function in human neural organoids. Semin Cell Dev Biol 2020; 111:52-59. [PMID: 32540123 DOI: 10.1016/j.semcdb.2020.05.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/17/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Over the last decade, scientists have begun to model CNS development, function, and disease in vitro using human pluripotent stem cell (hPSC)-derived organoids. Using traditional protocols, these 3D tissues are generated by combining the innate emergent properties of differentiating hPSC aggregates with a bioreactor environment that induces interstitial transport of oxygen and nutrients and an optional supportive hydrogel extracellular matrix (ECM). During extended culture, the hPSC-derived neural organoids (hNOs) obtain millimeter scale sizes with internal microscale cytoarchitectures, cellular phenotypes, and neuronal circuit behaviors mimetic of those observed in the developing brain, eye, or spinal cord. Early studies evaluated the cytoarchitectural and phenotypical character of these organoids and provided unprecedented insight into the morphogenetic processes that govern CNS development. Comparisons to human fetal tissues revealed their significant similarities and differences. While hNOs have current disease modeling applications and significant future promise, their value as anatomical and physiological models is limited because they fail to form reproducibly and recapitulate more mature in vivo features. These include biomimetic macroscale tissue morphology, positioning of morphogen signaling centers to orchestrate appropriate spatial organization and intra- and inter-connectivity of discrete tissue regions, maturation of physiologically relevant neural circuits, and formation of vascular networks that can support sustained in vitro tissue growth. To address these inadequacies scientists have begun to integrate organoid culture with bioengineering techniques and methodologies including genome editing, biomaterials, and microfabricated and microfluidic platforms that enable spatiotemporal control of cellular differentiation or the biochemical and biophysical cues that orchestrate organoid morphogenesis. This review will examine recent advances in hNO technologies and culture strategies that promote reproducible in vitro morphogenesis and greater biomimicry in structure and function.
Collapse
Affiliation(s)
- Nikolai J Fedorchak
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Nisha Iyer
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Randolph S Ashton
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, United States; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, United States.
| |
Collapse
|
73
|
Wang M, Yu Z, Li G, Yu X. Multiple Morphological Factors Underlie Experience-Dependent Cross-Modal Plasticity in the Developing Sensory Cortices. Cereb Cortex 2020; 30:2418-2433. [PMID: 31828301 DOI: 10.1093/cercor/bhz248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/21/2019] [Accepted: 09/18/2019] [Indexed: 11/14/2022] Open
Abstract
Sensory experience regulates the structural and functional wiring of sensory cortices. In previous work, we showed that whisker deprivation (WD) from birth not only reduced excitatory synaptic transmission of layer (L) 2/3 pyramidal neurons of the correspondent barrel cortex in mice, but also cross-modally reduced synaptic transmission of L2/3 pyramidal neurons in other sensory cortices. Here, we used in utero electroporation, in combination with optical clearing, to examine the main morphological components regulating neural circuit wiring, namely presynaptic bouton density, spine density, as well as dendrite and axon arbor lengths. We found that WD from P0 to P14 reduced presynaptic bouton density in both L4 and L2/3 inputs to L2/3 pyramidal neurons, as well as spine density across the dendritic tree of L2/3 pyramidal neurons, in the barrel field of the primary somatosensory cortex. The cross-modal effects in the primary auditory cortex were manifested mostly as reduced dendrite and axon arbor size, as well as reduced bouton density of L2/3 inputs. Increasing sensory experience by rearing mice in an enriched environment rescued the effects of WD. Together, these results demonstrate that multiple morphological factors contribute to experience-dependent structural plasticity during early wiring of the sensory cortices.
Collapse
Affiliation(s)
- Miao Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zixian Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangying Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
74
|
Kurihara T, Kouyama-Suzuki E, Satoga M, Li X, Badawi M, Thiha, Baig DN, Yanagawa T, Uemura T, Mori T, Tabuchi K. DNA repair protein RAD51 enhances the CRISPR/Cas9-mediated knock-in efficiency in brain neurons. Biochem Biophys Res Commun 2020; 524:621-628. [PMID: 32029273 DOI: 10.1016/j.bbrc.2020.01.132] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 10/25/2022]
Abstract
Gene knock-in using the CRISPR/Cas9 system can be achieved in a specific population of neurons in the mouse brain, by using in utero electroporation to introduce DNA fragments into neural progenitor cells. Using this strategy, we previously knocked-in the EGFP coding sequence into the N-terminal region of the β-actin gene specifically in the pyramidal neurons in layer 2/3 of the somatosensory cortex. However, the knock-in efficiency was less than 2% of the transfected neurons. In this study, we sought to improve the knock-in efficiency using this system. First, we varied the length of the homology arms of the β-actin donor template DNA, and found that the knock-in efficiency was increased to ∼14% by extending the length of the 5' and 3' homology arms to 1.6 kb and 2.0 kb, respectively. We then tested the effect of the DNA repair protein RAD51 and the knock-in efficiency was increased up to 2.5-fold when co-transfecting with two different β-actin and a camk2a targeting EGFP knock-in modules. The RAD51 overexpression did not alter the migration of developing neurons, density or morphology of the dendritic spines compared to those in neurons not transfected with RAD51. RAD51 expression will be useful for increasing the knock-in efficiency in neurons in vivo by CRISPR/Cas9-mediated homology directed repair (HDR).
Collapse
Affiliation(s)
- Taiga Kurihara
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan
| | - Emi Kouyama-Suzuki
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan
| | - Michiru Satoga
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan
| | - Xue Li
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan
| | - Moataz Badawi
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan
| | - Thiha
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan
| | - Deeba Noreen Baig
- School of Life Sciences Forman Christian College (A Chartared University), Lahore, 54600, Pakistan
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Takeshi Uemura
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan
| | - Takuma Mori
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, 390-8621, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan.
| |
Collapse
|
75
|
An early phase of instructive plasticity before the typical onset of sensory experience. Nat Commun 2020; 11:11. [PMID: 31896763 PMCID: PMC6940391 DOI: 10.1038/s41467-019-13872-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 12/04/2019] [Indexed: 11/09/2022] Open
Abstract
While early experience with moving stimuli is necessary for the development of direction selectivity in visual cortex of carnivores, it is unclear whether experience exerts a permissive or instructive influence. To test if the specific parameters of the experienced stimuli could instructively sculpt the emergent responses, visually naive ferrets were exposed to several hours of experience with unusual spatiotemporal patterns. In the most immature ferrets, cortical neurons developed selectivity to these patterns, indicating an instructive influence. In animals that were 1–10 days more mature, exposure to the same patterns led to a developmentally-typical increase in direction selectivity. We conclude that visual development progresses via an early phase of instructive plasticity, when the specific patterns of neural activity shape the specific parameters of the emerging response properties, followed by a late phase of permissive maturation, when sensory-driven activity merely serves to enhance the response properties already seeded in cortical circuits. Brain circuits exhibit different amounts of plasticity over different developmental stages. Here authors show that there is a transition of the influence of spatiotemporal patterns, from instructive to permissive, around the time of the onset of visual experience.
Collapse
|
76
|
Abstract
In utero electroporation is a rapid and powerful technique to study the development of many brain regions. This approach presents several advantages over other methods to study specific steps of brain development in vivo, from proliferation to synaptic integration. Here, we describe in detail the individual steps necessary to carry out the technique. We also highlight the variations that can be implemented to target different cerebral structures and to study specific steps of development.
Collapse
|
77
|
Saito K, Okamoto M, Watanabe Y, Noguchi N, Nagasaka A, Nishina Y, Shinoda T, Sakakibara A, Miyata T. Dorsal-to-Ventral Cortical Expansion Is Physically Primed by Ventral Streaming of Early Embryonic Preplate Neurons. Cell Rep 2019; 29:1555-1567.e5. [DOI: 10.1016/j.celrep.2019.09.075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/11/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023] Open
|
78
|
Arcaro MJ, Schade PF, Livingstone MS. Universal Mechanisms and the Development of the Face Network: What You See Is What You Get. Annu Rev Vis Sci 2019; 5:341-372. [PMID: 31226011 PMCID: PMC7568401 DOI: 10.1146/annurev-vision-091718-014917] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Our assignment was to review the development of the face-processing network, an assignment that carries the presupposition that a face-specific developmental program exists. We hope to cast some doubt on this assumption and instead argue that the development of face processing is guided by the same ubiquitous rules that guide the development of cortex in general.
Collapse
Affiliation(s)
- Michael J Arcaro
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Peter F Schade
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | | |
Collapse
|
79
|
Konno D, Kishida C, Maehara K, Ohkawa Y, Kiyonari H, Okada S, Matsuzaki F. Dmrt factors determine the positional information of cerebral cortical progenitors via differential suppression of homeobox genes. Development 2019; 146:dev.174243. [PMID: 31371378 DOI: 10.1242/dev.174243] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 07/23/2019] [Indexed: 01/06/2023]
Abstract
The spatiotemporal identity of neural progenitors and the regional control of neurogenesis are essential for the development of cerebral cortical architecture. Here, we report that mammalian DM domain factors (Dmrt) determine the identity of cerebral cortical progenitors. Among the Dmrt family genes expressed in the developing dorsal telencephalon, Dmrt3 and Dmrta2 show a medialhigh/laterallow expression gradient. Their simultaneous loss confers a ventral identity to dorsal progenitors, resulting in the ectopic expression of Gsx2 and massive production of GABAergic olfactory bulb interneurons in the dorsal telencephalon. Furthermore, double-mutant progenitors in the medial region exhibit upregulated Pax6 and more lateral characteristics. These ventral and lateral shifts in progenitor identity depend on Dmrt gene dosage. We also found that Dmrt factors bind to Gsx2 and Pax6 enhancers to suppress their expression. Our findings thus reveal that the graded expression of Dmrt factors provide positional information for progenitors by differentially repressing downstream genes in the developing cerebral cortex.
Collapse
Affiliation(s)
- Daijiro Konno
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan .,Division of Pathophysiology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Chiaki Kishida
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Kazumitsu Maehara
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroshi Kiyonari
- Laboratories for Animal Resource Development and Genetic Engineering (LARGE), RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Seiji Okada
- Division of Pathophysiology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
80
|
Abstract
Tactile sensory information from facial whiskers provides nocturnal tunnel-dwelling rodents, including mice and rats, with important spatial and textural information about their immediate surroundings. Whiskers are moved back and forth to scan the environment (whisking), and touch signals from each whisker evoke sparse patterns of neuronal activity in whisker-related primary somatosensory cortex (wS1; barrel cortex). Whisking is accompanied by desynchronized brain states and cell-type-specific changes in spontaneous and evoked neuronal activity. Tactile information, including object texture and location, appears to be computed in wS1 through integration of motor and sensory signals. wS1 also directly controls whisker movements and contributes to learned, whisker-dependent, goal-directed behaviours. The cell-type-specific neuronal circuitry in wS1 that contributes to whisker sensory perception is beginning to be defined.
Collapse
|
81
|
Linscott ML, Chung WCJ. TET1 regulates fibroblast growth factor 8 transcription in gonadotropin releasing hormone neurons. PLoS One 2019; 14:e0220530. [PMID: 31361780 PMCID: PMC6667164 DOI: 10.1371/journal.pone.0220530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022] Open
Abstract
Fibroblast growth factor 8 (FGF8) is a potent morphogen that regulates the ontogenesis of gonadotropin-releasing hormone (GnRH) neurons, which control the hypothalamus-pituitary-gonadal (HPG) axis, and therefore reproductive success. Indeed, FGF8 and FGFR1 deficiency severely compromises vertebrate reproduction in mice and humans and is associated with Kallmann Syndrome (KS), a congenital disease characterized by hypogonadotropic hypogonadism associated with anosmia. Our laboratory demonstrated that FGF8 signaling through FGFR1, both of which are KS-related genes, is necessary for proper GnRH neuron development in mice and humans. Here, we investigated the possibility that non-genetic factors, such as the epigenome, may contribute to KS onset. For this purpose, we developed an embryonic explant model, utilizing the mouse olfactory placode (OP), the birthplace of GnRH neurons. We show that TET1, which converts 5-methylcytosine residues (5mC) to 5-hydroxymethylated cytosines (5hmC), controls transcription of Fgf8 during GnRH neuron ontogenesis. Through MeDIP and ChIP RT-qPCR we found that TET1 bound to specific CpG islands on the Fgf8 promoter. We found that the temporal expression of Fgf8 correlates with not only TET1 binding, but also with 5hmC enrichment. siRNA knockdown of Tet1 reduced Fgf8 and Fgfr1 mRNA expression. During this time period, Fgf8 also switched histone status, most likely via recruitment of EZH2, a major component of the polycomb repressor complex-2 (PRC2) at E13.5. Together, these studies underscore the significance of epigenetics and chromatin modifications to temporally regulated genes involved in KS.
Collapse
Affiliation(s)
- Megan L. Linscott
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Wilson C. J. Chung
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
- School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| |
Collapse
|
82
|
FGF Signaling Directs the Cell Fate Switch from Neurons to Astrocytes in the Developing Mouse Cerebral Cortex. J Neurosci 2019; 39:6081-6094. [PMID: 31175212 DOI: 10.1523/jneurosci.2195-18.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 05/09/2019] [Accepted: 05/17/2019] [Indexed: 11/21/2022] Open
Abstract
During mammalian neocortical development, neural precursor cells generate neurons first and astrocytes later. The cell fate switch from neurons to astrocytes is a key process generating proper numbers of neurons and astrocytes. Although the intracellular mechanisms regulating this cell fate switch have been well characterized, extracellular regulators are still largely unknown. Here, we uncovered that fibroblast growth factor (FGF) regulates the cell fate switch from neurons to astrocytes in the developing cerebral cortex using mice of both sexes. We found that the FGF signaling pathway is activated in radial glial cells of the ventricular zone at time points corresponding to the switch in cell fate. Our loss- and gain-of-function studies using in utero electroporation indicate that activation of FGF signaling is necessary and sufficient to change cell fates from neurons to astrocytes. We further found that the FGF-induced neuron-astrocyte cell fate switch is mediated by the MAPK pathway. These results indicate that FGF is a critical extracellular regulator of the cell fate switch from neurons to astrocytes in the mammalian cerebral cortex.SIGNIFICANCE STATEMENT Although the intracellular mechanisms regulating the neuron-astrocyte cell fate switch in the mammalian cerebral cortex during development have been well studied, their upstream extracellular regulators remain unknown. By using in utero electroporation, our study provides in vivo data showing that activation of FGF signaling is necessary and sufficient for changing cell fates from neurons to astrocytes. Manipulation of FGF signaling activity led to drastic changes in the numbers of neurons and astrocytes. These results indicate that FGF is a key extracellular regulator determining the numbers of neurons and astrocytes in the mammalian cerebral cortex, and is indispensable for the establishment of appropriate neural circuitry.
Collapse
|
83
|
Shu P, Wu C, Ruan X, Liu W, Hou L, Fu H, Wang M, Liu C, Zeng Y, Chen P, Yin B, Yuan J, Qiang B, Peng X, Zhong W. Opposing Gradients of MicroRNA Expression Temporally Pattern Layer Formation in the Developing Neocortex. Dev Cell 2019; 49:764-785.e4. [PMID: 31080058 DOI: 10.1016/j.devcel.2019.04.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 11/08/2018] [Accepted: 04/07/2019] [Indexed: 12/24/2022]
Abstract
The precisely timed generation of different neuronal types is a hallmark of development from invertebrates to vertebrates. In the developing mammalian neocortex, neural stem cells change competence over time to sequentially produce six layers of functionally distinct neurons. Here, we report that microRNAs (miRNAs) are dispensable for stem-cell self-renewal and neuron production but essential for timing neocortical layer formation and specifying laminar fates. Specifically, as neurogenesis progresses, stem cells reduce miR-128 expression and miR-9 activity but steadily increase let-7 expression, whereas neurons initially maintain the differences in miRNA expression present at birth. Moreover, miR-128, miR-9, and let-7 are functionally distinct; capable of specifying neurons for layer VI and layer V and layers IV, III, and II, respectively; and transiently altering their relative levels of expression can modulate stem-cell competence in a neurogenic-stage-specific manner to shift neuron production between earlier-born and later-born fates, partly by temporally regulating a neurogenesis program involving Hmga2.
Collapse
Affiliation(s)
- Pengcheng Shu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chao Wu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiangbin Ruan
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Wei Liu
- Department of Anatomy and Histology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Lin Hou
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Hongye Fu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Ming Wang
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chang Liu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yi Zeng
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Pan Chen
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Bin Yin
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Jiangang Yuan
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Boqin Qiang
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiaozhong Peng
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| | - Weimin Zhong
- Department of Molecular, Cellular, and Developmental Biology, Yale University, P.O. Box 208103, New Haven, CT 06520, USA.
| |
Collapse
|
84
|
De Clercq S, Keruzore M, Desmaris E, Pollart C, Assimacopoulos S, Preillon J, Ascenzo S, Matson CK, Lee M, Nan X, Li M, Nakagawa Y, Hochepied T, Zarkower D, Grove EA, Bellefroid EJ. DMRT5 Together with DMRT3 Directly Controls Hippocampus Development and Neocortical Area Map Formation. Cereb Cortex 2019; 28:493-509. [PMID: 28031177 DOI: 10.1093/cercor/bhw384] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/16/2016] [Indexed: 11/14/2022] Open
Abstract
Mice that are constitutively null for the zinc finger doublesex and mab-3 related (Dmrt) gene, Dmrt5/Dmrta2, show a variety of patterning abnormalities in the cerebral cortex, including the loss of the cortical hem, a powerful cortical signaling center. In conditional Dmrt5 gain of function and loss of function mouse models, we generated bidirectional changes in the neocortical area map without affecting the hem. Analysis indicated that DMRT5, independent of the hem, directs the rostral-to-caudal pattern of the neocortical area map. Thus, DMRT5 joins a small number of transcription factors shown to control directly area size and position in the neocortex. Dmrt5 deletion after hem formation also reduced hippocampal size and shifted the position of the neocortical/paleocortical boundary. Dmrt3, like Dmrt5, is expressed in a gradient across the cortical primordium. Mice lacking Dmrt3 show cortical patterning defects akin to but milder than those in Dmrt5 mutants, perhaps in part because Dmrt5 expression increases in the absence of Dmrt3. DMRT5 upregulates Dmrt3 expression and negatively regulates its own expression, which may stabilize the level of DMRT5. Together, our findings indicate that finely tuned levels of DMRT5, together with DMRT3, regulate patterning of the cerebral cortex.
Collapse
Affiliation(s)
- Sarah De Clercq
- ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Marc Keruzore
- ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Elodie Desmaris
- ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Charlotte Pollart
- ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | | | - Julie Preillon
- ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Sabrina Ascenzo
- ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Clinton K Matson
- Department of Genetics, Cell Biology and Development , Minneapolis, MN 55455, USA
| | - Melody Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Xinsheng Nan
- School of Medicine and School of Bioscience , Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF10 3XQ, UK
| | - Meng Li
- School of Medicine and School of Bioscience , Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF10 3XQ, UK
| | - Yasushi Nakagawa
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tino Hochepied
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.,Inflammation Research Center, VIB, B-9052 Ghent, Belgium
| | - David Zarkower
- Department of Genetics, Cell Biology and Development , Minneapolis, MN 55455, USA
| | - Elizabeth A Grove
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Eric J Bellefroid
- ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| |
Collapse
|
85
|
Paquola C, Vos De Wael R, Wagstyl K, Bethlehem RAI, Hong SJ, Seidlitz J, Bullmore ET, Evans AC, Misic B, Margulies DS, Smallwood J, Bernhardt BC. Microstructural and functional gradients are increasingly dissociated in transmodal cortices. PLoS Biol 2019; 17:e3000284. [PMID: 31107870 PMCID: PMC6544318 DOI: 10.1371/journal.pbio.3000284] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/31/2019] [Accepted: 05/08/2019] [Indexed: 01/10/2023] Open
Abstract
While the role of cortical microstructure in organising neural function is well established, it remains unclear how structural constraints can give rise to more flexible elements of cognition. While nonhuman primate research has demonstrated a close structure-function correspondence, the relationship between microstructure and function remains poorly understood in humans, in part because of the reliance on post mortem analyses, which cannot be directly related to functional data. To overcome this barrier, we developed a novel approach to model the similarity of microstructural profiles sampled in the direction of cortical columns. Our approach was initially formulated based on an ultra-high-resolution 3D histological reconstruction of an entire human brain and then translated to myelin-sensitive magnetic resonance imaging (MRI) data in a large cohort of healthy adults. This novel method identified a system-level gradient of microstructural differentiation traversing from primary sensory to limbic regions that followed shifts in laminar differentiation and cytoarchitectural complexity. Importantly, while microstructural and functional gradients described a similar hierarchy, they became increasingly dissociated in transmodal default mode and fronto-parietal networks. Meta-analytic decoding of these topographic dissociations highlighted involvement in higher-level aspects of cognition, such as cognitive control and social cognition. Our findings demonstrate a relative decoupling of macroscale functional from microstructural gradients in transmodal regions, which likely contributes to the flexible role these regions play in human cognition.
Collapse
Affiliation(s)
- Casey Paquola
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Reinder Vos De Wael
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Konrad Wagstyl
- McGill Centre for Integrative Neuroscience, McGill University, Montreal, Canada
| | - Richard A. I. Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Seok-Jun Hong
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Jakob Seidlitz
- Developmental Neurogenomics Unit, National Institute of Mental Health, Bethesda, Maryland, United States of America
- Brain Mapping Unit, University of Cambridge, Department of Psychiatry, Cambridge, United Kingdom
| | - Edward T. Bullmore
- Brain Mapping Unit, University of Cambridge, Department of Psychiatry, Cambridge, United Kingdom
| | - Alan C. Evans
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
- McGill Centre for Integrative Neuroscience, McGill University, Montreal, Canada
| | - Bratislav Misic
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | | | | | - Boris C. Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| |
Collapse
|
86
|
Nakagawa Y. Development of the thalamus: From early patterning to regulation of cortical functions. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e345. [PMID: 31034163 DOI: 10.1002/wdev.345] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
The thalamus is a brain structure of the vertebrate diencephalon that plays a central role in regulating diverse functions of the cerebral cortex. In traditional view of vertebrate neuroanatomy, the thalamus includes three regions, dorsal thalamus, ventral thalamus, and epithalamus. Recent molecular embryological studies have redefined the thalamus and the associated axial nomenclature of the diencephalon in the context of forebrain patterning. This new view has provided a useful conceptual framework for studies on molecular mechanisms of patterning, neurogenesis and fate specification in the thalamus as well as the guidance mechanisms for thalamocortical axons. Additionally, the availability of genetic tools in mice has led to important findings on how thalamic development is linked to the development of other brain regions, particularly the cerebral cortex. This article will give an overview of the organization of the embryonic thalamus and how progenitor cells in the thalamus generate neurons that are organized into discrete nuclei. I will then discuss how thalamic development is orchestrated with the development of the cerebral cortex and other brain regions. This article is categorized under: Nervous System Development > Vertebrates: Regional Development Nervous System Development > Vertebrates: General Principles.
Collapse
Affiliation(s)
- Yasushi Nakagawa
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
87
|
Chan C, Kamiguchi H, Shimogori T. Spatially restricted long-term transgene expression in the developing skin used for studying the interaction of epidermal development and sensory innervation. Dev Growth Differ 2019; 61:276-282. [PMID: 30968390 DOI: 10.1111/dgd.12603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 11/30/2022]
Abstract
Skin development is tightly temporally coordinated with its sensory innervation, which consists of the peripheral branches of the dorsal root ganglion (DRG) axons. Various studies suggest that the skin produces a long-range attractant for the sensory axons. However, the exact identity of the guidance cue(s) remains unclear. To reveal the detailed molecular mechanism that controls DRG axon guidance and targeting, manipulation of specific skin layers at specific time points are required. To test a variety of attractants that can be expressed in specific skin layers at specific timepoints, we combined in utero electroporation with the Tol2 transposon system to induce long-term transgene expression in the developing mouse skin, including in the highly proliferative epidermal stem cells (basal layer) and their descendants (spinous and granular layer cells). The plasmid solution was injected as close to the hindpaw plantar surface as possible. Immediately, electric pulses were passed through the embryo to transduce the plasmid DNA into hindpaw skin cells. Balancing outcome measurements including: embryo survival, transfection efficiency, and the efficiency of transgene integration into host cells, we found that IUE was best performed on E13.5, and using an electroporation voltage of 34V. After immunostaining embryonic and early postnatal skin tissue sections for keratinocyte and sensory axon markers, we observe the growth of axons into skin epidermal layers including areas expressing EGFP. Therefore, this method is useful for studying the interaction between axon growth and epidermal cell division/differentiation.
Collapse
Affiliation(s)
- Carmen Chan
- RIKEN Center for Brain Science, Lab for Neural Cell Dynamics, Wako, Saitama, Japan
| | - Hiroyuki Kamiguchi
- RIKEN Center for Brain Science, Lab for Neural Cell Dynamics, Wako, Saitama, Japan
| | - Tomomi Shimogori
- RIKEN Center for Brain Science, Lab for Molecular Mechanisms of Brain Development, Wako, Saitama, Japan
| |
Collapse
|
88
|
The Expression of Key Guidance Genes at a Forebrain Axon Turning Point Is Maintained by Distinct Fgfr Isoforms but a Common Downstream Signal Transduction Mechanism. eNeuro 2019; 6:eN-NWR-0086-19. [PMID: 30993182 PMCID: PMC6464512 DOI: 10.1523/eneuro.0086-19.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/24/2022] Open
Abstract
During development the axons of neurons grow toward and locate their synaptic partners to form functional neural circuits. Axons do so by reading a map of guidance cues expressed by surrounding tissues. Guidance cues are expressed at a precise space and time, but how guidance cue expression is regulated, and in a coordinated manner, is poorly understood. Semaphorins (Semas) and Slits are families of molecular ligands that guide axons. We showed previously that fibroblast growth factor (Fgf) signaling maintains sema3a and slit1 forebrain expression in Xenopus laevis, and these two repellents cooperate to guide retinal ganglion cell (RGC) axons away from the mid-diencephalon and on towards the optic tectum. Here, we investigate whether there are common features of the regulatory pathways that control the expression of these two guidance cues at this single axon guidance decision point. We isolated the sema3a proximal promoter and confirmed its responsiveness to Fgf signaling. Through misexpression of truncated Fgf receptors (Fgfrs), we found that sema3a forebrain expression is dependent on Fgfr2-4 but not Fgfr1. This is in contrast to slit1, whose expression we showed previously depends on Fgfr1 but not Fgfr2-4. Using pharmacological inhibitors and misexpression of constitutively active (CA) and dominant negative (DN) signaling intermediates, we find that while distinct Fgfrs regulate these two guidance genes, intracellular signaling downstream of Fgfrs appears to converge along the phosphoinositol 3-kinase (PI3K)-Akt signaling pathway. A common PI3K-Akt signaling pathway may allow for the coordinated expression of guidance cues that cooperate to direct axons at a guidance choice point.
Collapse
|
89
|
Kast RJ, Levitt P. Precision in the development of neocortical architecture: From progenitors to cortical networks. Prog Neurobiol 2019; 175:77-95. [PMID: 30677429 PMCID: PMC6402587 DOI: 10.1016/j.pneurobio.2019.01.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/02/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
Of all brain regions, the 6-layered neocortex has undergone the most dramatic changes in size and complexity during mammalian brain evolution. These changes, occurring in the context of a conserved set of organizational features that emerge through stereotypical developmental processes, are considered responsible for the cognitive capacities and sensory specializations represented within the mammalian clade. The modern experimental era of developmental neurobiology, spanning 6 decades, has deciphered a number of mechanisms responsible for producing the diversity of cortical neuron types, their precise connectivity and the role of gene by environment interactions. Here, experiments providing insight into the development of cortical projection neuron differentiation and connectivity are reviewed. This current perspective integrates discussion of classic studies and new findings, based on recent technical advances, to highlight an improved understanding of the neuronal complexity and precise connectivity of cortical circuitry. These descriptive advances bring new opportunities for studies related to the developmental origins of cortical circuits that will, in turn, improve the prospects of identifying pathogenic targets of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ryan J Kast
- Department of Pediatrics and Program in Developmental Neuroscience and Developmental Neurogenetics, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, USA
| | - Pat Levitt
- Department of Pediatrics and Program in Developmental Neuroscience and Developmental Neurogenetics, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, USA.
| |
Collapse
|
90
|
Huang Z, Wang Y. In Vivo Electroporation and Time-Lapse Imaging of the Rostral Migratory Stream in Developing Rodent Brain. ACTA ACUST UNITED AC 2019; 87:e65. [PMID: 30861320 DOI: 10.1002/cpns.65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interneurons in the olfactory bulb are generated from neuronal precursor cells migrating from the anterior subventricular zone (SVZa) throughout the embryonic and postnatal life of mammals. This article describes basic methods for in vivo electroporation to label SVZa cells of both embryonic and postnatal rats. In addition, it describes three methods for tracing SVZa progenitors and following their migration pathway and differentiation, including immunohistochemistry, time-lapse live imaging in slice culture, and time-lapse imaging following transplantation in slice culture. These methods may be applied to all strains of rats and mice, including reporter mice. They may also be combined with methods such as BrdU labeling, tamoxifen injection, and electrophysiology, allowing one to observe proliferation or control gene expression at specific times and for specific neuronal functions. With time-lapse live imaging, details of labeled cells can be studied, including morphology, motility pattern, differentiation, and crosstalk between cells. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Zhihui Huang
- Institute of Neuroscience, Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Ying Wang
- Institute of Clinical Research, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Department of Blood Transfusion, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
91
|
Jones WD, Guadiana SM, Grove EA. A model of neocortical area patterning in the lissencephalic mouse may hold for larger gyrencephalic brains. J Comp Neurol 2019; 527:1461-1477. [PMID: 30689213 DOI: 10.1002/cne.24643] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/21/2018] [Accepted: 01/02/2019] [Indexed: 12/14/2022]
Abstract
In the mouse, two telencephalic signaling centers orchestrate embryonic patterning of the cerebral cortex. From the rostral patterning center in the telencephalon, the Fibroblast Growth Factor, FGF8, disperses as a morphogen to establish the rostral to caudal axis of the neocortical area map. FGF8 coordinates with Wnt3a from the cortical hem to regulate graded expression of transcription factors that position neocortical areas, and control hippocampal development. Whether similar signaling centers pattern the much larger cortices of carnivore and primate species, however, is unclear. The limited dispersion range of FGF8 and Wnt3a is inconsistent with patterning larger cortical primordia. Yet the implication that different mechanisms organize cortex in different mammals flies in the face of the tenet that developmental patterning mechanisms are conserved across vertebrate species. In the present study, both signaling centers were identified in the ferret telencephalon, as were expression gradients of the patterning transcription factor genes regulated by FGF8 and Wnt3a. Notably, at the stage corresponding to the peak period of FGF8 signaling in the mouse neocortical primordium (NP), the NP was the same size in ferret and mouse, which would allow morphogen patterning of the ferret NP. Subsequently, the size of ferret neocortex shot past that of the mouse. Images from online databases further suggest that NP growth in humans, too, is slowed in early cortical development. We propose that if early growth in larger brains is held back, mechanisms that pattern the neocortical area map in the mouse could be conserved across mammalian species.
Collapse
Affiliation(s)
- William D Jones
- Department of Neurobiology, University of Chicago, Chicago, Illinois
| | - Sarah M Guadiana
- Department of Neurobiology, University of Chicago, Chicago, Illinois
| | - Elizabeth A Grove
- Department of Neurobiology, University of Chicago, Chicago, Illinois.,Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, Illinois.,Committee on Neurobiology, University of Chicago, Chicago, Illinois
| |
Collapse
|
92
|
Simi A, Studer M. Developmental genetic programs and activity-dependent mechanisms instruct neocortical area mapping. Curr Opin Neurobiol 2018; 53:96-102. [DOI: 10.1016/j.conb.2018.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/07/2018] [Accepted: 06/14/2018] [Indexed: 12/18/2022]
|
93
|
Embryonic Neocortical Microglia Express Toll-Like Receptor 9 and Respond to Plasmid DNA Injected into the Ventricle: Technical Considerations Regarding Microglial Distribution in Electroporated Brain Walls. eNeuro 2018; 5:eN-MNT-0312-18. [PMID: 30627652 PMCID: PMC6325556 DOI: 10.1523/eneuro.0312-18.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/19/2018] [Accepted: 10/27/2018] [Indexed: 12/25/2022] Open
Abstract
Microglia, the resident immune cells in the CNS, play multiple roles during development. In the embryonic cerebral wall, microglia modulate the functions of neural stem/progenitor cells through their distribution in regions undergoing cell proliferation and/or differentiation. Previous studies using CX3CR1-GFP transgenic mice demonstrated that microglia extensively survey these regions. To simultaneously visualize microglia and neural-lineage cells that interact with each other, we applied the in utero electroporation (IUE) technique, which has been widely used for gene-transfer in neurodevelopmental studies, to CX3CR1-GFP mice (males and females). However, we unexpectedly faced a technical problem: although microglia are normally distributed homogeneously throughout the mid-embryonic cortical wall with only limited luminal entry, the intraventricular presence of exogenously derived plasmid DNAs induced microglia to accumulate along the apical surface of the cortex and aggregate in the choroid plexus. This effect was independent of capillary needle puncture of the brain wall or application of electrical pulses. The microglial response occurred at plasmid DNA concentrations lower than those routinely used for IUE, and was mediated by activation of Toll-like receptor 9 (TLR9), an innate immune sensor that recognizes unmethylated cytosine-phosphate guanosine motifs abundant in microbial DNA. Administration of plasmid DNA together with oligonucleotide 2088, the antagonist of TLR9, partially restored the dispersed intramural localization of microglia and significantly decreased luminal accumulation of these cells. Thus, via TLR9, intraventricular plasmid DNA administration causes aberrant distribution of embryonic microglia, suggesting that the behavior of microglia in brain primordia subjected to IUE should be carefully interpreted.
Collapse
|
94
|
Hatami M, Conrad S, Naghsh P, Alvarez-Bolado G, Skutella T. Cell-Biological Requirements for the Generation of Dentate Gyrus Granule Neurons. Front Cell Neurosci 2018; 12:402. [PMID: 30483057 PMCID: PMC6240695 DOI: 10.3389/fncel.2018.00402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/18/2018] [Indexed: 12/22/2022] Open
Abstract
The dentate gyrus (DG) receives highly processed information from the associative cortices functionally integrated in the trisynaptic hippocampal circuit, which contributes to the formation of new episodic memories and the spontaneous exploration of novel environments. Remarkably, the DG is the only brain region currently known to have high rates of neurogenesis in adults (Andersen et al., 1966, 1971). The DG is involved in several neurodegenerative disorders, including clinical dementia, schizophrenia, depression, bipolar disorder and temporal lobe epilepsy. The principal neurons of the DG are the granule cells. DG granule cells generated in culture would be an ideal model to investigate their normal development and the causes of the pathologies in which they are involved and as well as possible therapies. Essential to establish such in vitro models is the precise definition of the most important cell-biological requirements for the differentiation of DG granule cells. This requires a deeper understanding of the precise molecular and functional attributes of the DG granule cells in vivo as well as the DG cells derived in vitro. In this review we outline the neuroanatomical, molecular and cell-biological components of the granule cell differentiation pathway, including some growth- and transcription factors essential for their development. We summarize the functional characteristics of DG granule neurons, including the electrophysiological features of immature and mature granule cells and the axonal pathfinding characteristics of DG neurons. Additionally, we discuss landmark studies on the generation of dorsal telencephalic precursors from pluripotent stem cells (PSCs) as well as DG neuron differentiation in culture. Finally, we provide an outlook and comment critical aspects.
Collapse
Affiliation(s)
- Maryam Hatami
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Pooyan Naghsh
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | | | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
95
|
Metabolic regulation and glucose sensitivity of cortical radial glial cells. Proc Natl Acad Sci U S A 2018; 115:10142-10147. [PMID: 30224493 DOI: 10.1073/pnas.1808066115] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The primary stem cells of the cerebral cortex are the radial glial cells (RGCs), and disturbances in their operation lead to myriad brain disorders in all mammals from mice to humans. Here, we found in mice that maternal gestational obesity and hyperglycemia can impair the maturation of RGC fibers and delay cortical neurogenesis. To investigate potential mechanisms, we used optogenetic live-imaging approaches in embryonic cortical slices. We found that Ca2+ signaling regulates mitochondrial transport and is crucial for metabolic support in RGC fibers. Cyclic intracellular Ca2+ discharge from localized RGC fiber segments detains passing mitochondria and ensures their proper distribution and enrichment at specific sites such as endfeet. Impairment of mitochondrial function caused an acute loss of Ca2+ signaling, while hyperglycemia decreased Ca2+ activity and impaired mitochondrial transport, leading to degradation of the RGC scaffold. Our findings uncover a physiological mechanism indicating pathways by which gestational metabolic disturbances can interfere with brain development.
Collapse
|
96
|
Kawasaki H. Molecular Investigations of the Development and Diseases of Cerebral Cortex Folding using Gyrencephalic Mammal Ferrets. Biol Pharm Bull 2018; 41:1324-1329. [DOI: 10.1248/bpb.b18-00142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University
| |
Collapse
|
97
|
Ricard C, Arroyo ED, He CX, Portera-Cailliau C, Lepousez G, Canepari M, Fiole D. Two-photon probes for in vivo multicolor microscopy of the structure and signals of brain cells. Brain Struct Funct 2018; 223:3011-3043. [PMID: 29748872 PMCID: PMC6119111 DOI: 10.1007/s00429-018-1678-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 05/03/2018] [Indexed: 02/07/2023]
Abstract
Imaging the brain of living laboratory animals at a microscopic scale can be achieved by two-photon microscopy thanks to the high penetrability and low phototoxicity of the excitation wavelengths used. However, knowledge of the two-photon spectral properties of the myriad fluorescent probes is generally scarce and, for many, non-existent. In addition, the use of different measurement units in published reports further hinders the design of a comprehensive imaging experiment. In this review, we compile and homogenize the two-photon spectral properties of 280 fluorescent probes. We provide practical data, including the wavelengths for optimal two-photon excitation, the peak values of two-photon action cross section or molecular brightness, and the emission ranges. Beyond the spectroscopic description of these fluorophores, we discuss their binding to biological targets. This specificity allows in vivo imaging of cells, their processes, and even organelles and other subcellular structures in the brain. In addition to probes that monitor endogenous cell metabolism, studies of healthy and diseased brain benefit from the specific binding of certain probes to pathology-specific features, ranging from amyloid-β plaques to the autofluorescence of certain antibiotics. A special focus is placed on functional in vivo imaging using two-photon probes that sense specific ions or membrane potential, and that may be combined with optogenetic actuators. Being closely linked to their use, we examine the different routes of intravital delivery of these fluorescent probes according to the target. Finally, we discuss different approaches, strategies, and prerequisites for two-photon multicolor experiments in the brains of living laboratory animals.
Collapse
Affiliation(s)
- Clément Ricard
- Brain Physiology Laboratory, CNRS UMR 8118, 75006, Paris, France
- Faculté de Sciences Fondamentales et Biomédicales, Université Paris Descartes, PRES Sorbonne Paris Cité, 75006, Paris, France
- Fédération de Recherche en Neurosciences FR 3636, Paris, 75006, France
| | - Erica D Arroyo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Cynthia X He
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Gabriel Lepousez
- Unité Perception et Mémoire, Département de Neuroscience, Institut Pasteur, 25 rue du Docteur Roux, 75724, Paris Cedex 15, France
| | - Marco Canepari
- Laboratory for Interdisciplinary Physics, UMR 5588 CNRS and Université Grenoble Alpes, 38402, Saint Martin d'Hères, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Grenoble, France
- Institut National de la Santé et Recherche Médicale (INSERM), Grenoble, France
| | - Daniel Fiole
- Unité Biothérapies anti-Infectieuses et Immunité, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, BP 73, 91223, Brétigny-sur-Orge cedex, France.
- Human Histopathology and Animal Models, Infection and Epidemiology Department, Institut Pasteur, 28 rue du docteur Roux, 75725, Paris Cedex 15, France.
- ESRF-The European Synchrotron, 38043, Grenoble cedex, France.
| |
Collapse
|
98
|
Cwetsch AW, Pinto B, Savardi A, Cancedda L. In vivo methods for acute modulation of gene expression in the central nervous system. Prog Neurobiol 2018; 168:69-85. [PMID: 29694844 PMCID: PMC6080705 DOI: 10.1016/j.pneurobio.2018.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Abstract
Accurate and timely expression of specific genes guarantees the healthy development and function of the brain. Indeed, variations in the correct amount or timing of gene expression lead to improper development and/or pathological conditions. Almost forty years after the first successful gene transfection in in vitro cell cultures, it is currently possible to regulate gene expression in an area-specific manner at any step of central nervous system development and in adulthood in experimental animals in vivo, even overcoming the very poor accessibility of the brain. Here, we will review the diverse approaches for acute gene transfer in vivo, highlighting their advantages and disadvantages with respect to the efficiency and specificity of transfection as well as to brain accessibility. In particular, we will present well-established chemical, physical and virus-based approaches suitable for different animal models, pointing out their current and future possible applications in basic and translational research as well as in gene therapy.
Collapse
Affiliation(s)
- Andrzej W Cwetsch
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126, Pisa, Italy
| | - Annalisa Savardi
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; DulbeccoTelethon Institute, Italy.
| |
Collapse
|
99
|
Terrigno M, Bertacchi M, Pandolfini L, Baumgart M, Calvello M, Cellerino A, Studer M, Cremisi F. The microRNA miR-21 Is a Mediator of FGF8 Action on Cortical COUP-TFI Translation. Stem Cell Reports 2018; 11:756-769. [PMID: 30174317 PMCID: PMC6135738 DOI: 10.1016/j.stemcr.2018.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
The morphogen FGF8 plays a pivotal role in neocortical area patterning through its inhibitory effect on COUP-TFI/Nr2f1 anterior expression, but its mechanism of action is poorly understood. We established an in vitro model of mouse embryonic stem cell corticogenesis in which COUP-TFI protein expression is inhibited by the activation of FGF8 in a time window corresponding to cortical area patterning. Interestingly, overexpression of the COUP-TFI 3'UTR reduces the inhibitory effect of FGF8 on COUP-TFI translation. FGF8 induces the expression of few miRNAs targeting COUP-TFI 3'UTR in silico. We found that the functional inhibition of miR-21 can effectively counteract the inhibitory effect of FGF8 in vitro and regulate COUP-TFI protein levels in vivo. Accordingly, miR-21 expression is complementary to COUP-TFI expression during corticogenesis. These data support a translational control of COUP-TFI gradient expression by FGF8 via miR-21 and contribute to our understanding of how regionalized expression is established during neocortical area mapping.
Collapse
Affiliation(s)
- Marco Terrigno
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy
| | | | - Luca Pandolfini
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy
| | - Mario Baumgart
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | | | - Alessandro Cellerino
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy; Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Michèle Studer
- Université Côte d'Azur (UCA), CNRS, Inserm, iBV, 06108 Nice, France
| | - Federico Cremisi
- Scuola Normale Superiore, Piazza dei Cavalieri, 7, Pisa 56126, Italy; Istituto di Biofisica CNR, Via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
100
|
Newman EA, Wu D, Taketo MM, Zhang J, Blackshaw S. Canonical Wnt signaling regulates patterning, differentiation and nucleogenesis in mouse hypothalamus and prethalamus. Dev Biol 2018; 442:236-248. [PMID: 30063881 DOI: 10.1016/j.ydbio.2018.07.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 01/10/2023]
Abstract
The hypothalamus is a small, but anatomically and functionally complex region of the brain whose development is poorly understood. In this study, we have explored its development by studying the canonical Wnt signaling pathway, generating gain and loss of function mutations of beta-catenin (Ctnnb1) in both hypothalamic and prethalamic neuroepithelium. Deletion of Ctnnb1 resulted in an anteriorized and hypoplastic hypothalamus. Posterior structures were lost or reduced, and anterior structures were expanded. In contrast, overexpression of a constitutively active mutant form of Ctnnb1 resulted in severe hyperplasia of prethalamus and hypothalamus, and expanded expression of a subset of posterior and premamillary hypothalamic markers. Moderate defects in differentiation of Arx-positive GABAergic neural precursors were observed in both prethalamus and hypothalamus of Ctnnb1 loss of function mutants, while in gain of function mutants, their differentiation was completely suppressed, although markers of prethalamic progenitors were preserved. Multiple other region-specific markers, including several specific posterior hypothalamic structures, were also suppressed in Ctnnb1 gain of function mutations. Severe, region-specific defects in hypothalamic nucleogenesis were also observed in both gain and loss of function mutations of Ctnnb1. Finally, both gain and loss of function of Ctnnb1 also produced severe, non-cell autonomous disruptions of pituitary development. These findings demonstrate a central and multifaceted role for canonical Wnt signaling in regulating growth, patterning, differentiation and nucleogenesis in multiple diencephalic regions.
Collapse
Affiliation(s)
- Elizabeth A Newman
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan Wu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jiangyang Zhang
- Department of Radiology, NYU Langone School of Medicine, New York, NY, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|