fbpx
Wikipedia

Development of the nervous system

The development of the nervous system, or neural development (neurodevelopment), refers to the processes that generate, shape, and reshape the nervous system of animals, from the earliest stages of embryonic development to adulthood. The field of neural development draws on both neuroscience and developmental biology to describe and provide insight into the cellular and molecular mechanisms by which complex nervous systems develop, from nematodes and fruit flies to mammals.

Defects in neural development can lead to malformations such as holoprosencephaly, and a wide variety of neurological disorders including limb paresis and paralysis, balance and vision disorders, and seizures,[1] and in humans other disorders such as Rett syndrome, Down syndrome and intellectual disability.[2]

Overview of vertebrate brain development Edit

 
Diagram of the vertebrate nervous system

The vertebrate central nervous system (CNS) is derived from the ectoderm—the outermost germ layer of the embryo. A part of the dorsal ectoderm becomes specified to neural ectoderm – neuroectoderm that forms the neural plate along the dorsal side of the embryo.[3] This is a part of the early patterning of the embryo (including the invertebrate embryo) that also establishes an anterior-posterior axis.[4][5] The neural plate is the source of the majority of neurons and glial cells of the CNS. The neural groove forms along the long axis of the neural plate, and the neural plate folds to give rise to the neural tube.[6] When the tube is closed at both ends it is filled with embryonic cerebrospinal fluid.[7] As the embryo develops, the anterior part of the neural tube expands and forms three primary brain vesicles, which become the forebrain (prosencephalon), midbrain (mesencephalon), and hindbrain (rhombencephalon). These simple, early vesicles enlarge and further divide into the telencephalon (future cerebral cortex and basal ganglia), diencephalon (future thalamus and hypothalamus), mesencephalon (future colliculi), metencephalon (future pons and cerebellum), and myelencephalon (future medulla).[8] The CSF-filled central chamber is continuous from the telencephalon to the central canal of the spinal cord, and constitutes the developing ventricular system of the CNS. Embryonic cerebrospinal fluid differs from that formed in later developmental stages, and from adult CSF; it influences the behavior of neural precursors.[7] Because the neural tube gives rise to the brain and spinal cord any mutations at this stage in development can lead to fatal deformities like anencephaly or lifelong disabilities like spina bifida. During this time, the walls of the neural tube contain neural stem cells, which drive brain growth as they divide many times. Gradually some of the cells stop dividing and differentiate into neurons and glial cells, which are the main cellular components of the CNS. The newly generated neurons migrate to different parts of the developing brain to self-organize into different brain structures. Once the neurons have reached their regional positions, they extend axons and dendrites, which allow them to communicate with other neurons via synapses. Synaptic communication between neurons leads to the establishment of functional neural circuits that mediate sensory and motor processing, and underlie behavior.[9]

 
Flowchart of human brain development

Aspects Edit

Some landmarks of neural development include the birth and differentiation of neurons from stem cell precursors, the migration of immature neurons from their birthplaces in the embryo to their final positions, outgrowth of axons and dendrites from neurons, guidance of the motile growth cone through the embryo towards postsynaptic partners, the generation of synapses between these axons and their postsynaptic partners, and finally the lifelong changes in synapses, which are thought to underlie learning and memory.

Typically, these neurodevelopmental processes can be broadly divided into two classes: activity-independent mechanisms and activity-dependent mechanisms. Activity-independent mechanisms are generally believed to occur as hardwired processes determined by genetic programs played out within individual neurons. These include differentiation, migration and axon guidance to their initial target areas. These processes are thought of as being independent of neural activity and sensory experience. Once axons reach their target areas, activity-dependent mechanisms come into play. Although synapse formation is an activity-independent event, modification of synapses and synapse elimination requires neural activity.

Developmental neuroscience uses a variety of animal models including the mouse Mus musculus, the fruit fly Drosophila melanogaster, the zebrafish Danio rerio, the frog Xenopus laevis, and the roundworm Caenorhabditis elegans.

Myelination, formation of the lipid myelin sheath around neuronal axons, is a process that is essential for normal brain function. The myelin sheath provides insulation for the nerve impulse when communicating between neural systems. Without it, the impulse would be disrupted and the signal would not reach its target, thus impairing normal functioning. Because so much of brain development occurs in the prenatal stage and infancy, it is crucial that myelination, along with cortical development occur properly. Magnetic resonance imaging (MRI) is a non-invasive technique used to investigate myelination and cortical maturation (the cortex is the outer layer of the brain composed of gray matter). Rather than showing the actual myelin, the MRI picks up on the myelin water fraction, a measure of myelin content. Multicomponent relaxometry (MCR) allow visualization and quantification of myelin content. MCR is also useful for tracking white matter maturation, which plays an important role in cognitive development. It has been discovered that in infancy, myelination occurs in a caudal–cranial, posterior-to-anterior pattern. Because there is little evidence of a relationship between myelination and cortical thickness, it was revealed that cortical thickness is independent of white matter. This allows various aspects of the brain to grow simultaneously, leading to a more fully developed brain.[10]

Neural induction Edit

During early embryonic development of the vertebrate, the dorsal ectoderm becomes specified to give rise to the epidermis and the nervous system; a part of the dorsal ectoderm becomes specified to neural ectoderm to form the neural plate which gives rise to the nervous system.[3][11] The conversion of undifferentiated ectoderm to neuroectoderm requires signals from the mesoderm. At the onset of gastrulation presumptive mesodermal cells move through the dorsal blastopore lip and form a layer of mesoderm in between the endoderm and the ectoderm. Mesodermal cells migrate along the dorsal midline to give rise to the notochord that develops into the vertebral column. Neuroectoderm overlying the notochord develops into the neural plate in response to a diffusible signal produced by the notochord. The remainder of the ectoderm gives rise to the epidermis. The ability of the mesoderm to convert the overlying ectoderm into neural tissue is called neural induction.

In the early embryo, the neural plate folds outwards to form the neural groove. Beginning in the future neck region, the neural folds of this groove close to create the neural tube. The formation of the neural tube from the ectoderm is called neurulation. The ventral part of the neural tube is called the basal plate; the dorsal part is called the alar plate. The hollow interior is called the neural canal, and the open ends of the neural tube, called the neuropores, close off.[12]

A transplanted blastopore lip can convert ectoderm into neural tissue and is said to have an inductive effect. Neural inducers are molecules that can induce the expression of neural genes in ectoderm explants without inducing mesodermal genes as well. Neural induction is often studied in Xenopus embryos since they have a simple body plan and there are good markers to distinguish between neural and non-neural tissue. Examples of neural inducers are the molecules noggin and chordin.

When embryonic ectodermal cells are cultured at low density in the absence of mesodermal cells they undergo neural differentiation (express neural genes), suggesting that neural differentiation is the default fate of ectodermal cells. In explant cultures (which allow direct cell-cell interactions) the same cells differentiate into epidermis. This is due to the action of BMP4 (a TGF-β family protein) that induces ectodermal cultures to differentiate into epidermis. During neural induction, noggin and chordin are produced by the dorsal mesoderm (notochord) and diffuse into the overlying ectoderm to inhibit the activity of BMP4. This inhibition of BMP4 causes the cells to differentiate into neural cells. Inhibition of TGF-β and BMP (bone morphogenetic protein) signaling can efficiently induce neural tissue from pluripotent stem cells.[13]

Regionalization Edit

In a later stage of development the superior part of the neural tube flexes at the level of the future midbrain—the mesencephalon, at the mesencephalic flexure or cephalic flexure. Above the mesencephalon is the prosencephalon (future forebrain) and beneath it is the rhombencephalon (future hindbrain).

The alar plate of the prosencephalon expands to form the telencephalon which gives rise to the cerebral hemispheres, whilst its basal plate becomes the diencephalon. The optical vesicle (which eventually become the optic nerve, retina and iris) forms at the basal plate of the prosencephalon.

Patterning of the nervous system Edit

In chordates, dorsal ectoderm forms all neural tissue and the nervous system. Patterning occurs due to specific environmental conditions - different concentrations of signaling molecules

Dorsoventral axis Edit

The ventral half of the neural plate is controlled by the notochord, which acts as the 'organiser'. The dorsal half is controlled by the ectoderm plate, which flanks either side of the neural plate.[14]

Ectoderm follows a default pathway to become neural tissue. Evidence for this comes from single, cultured cells of ectoderm, which go on to form neural tissue. This is postulated to be because of a lack of BMPs, which are blocked by the organiser. The organiser may produce molecules such as follistatin, noggin and chordin that inhibit BMPs.

The ventral neural tube is patterned by sonic hedgehog (Shh) from the notochord, which acts as the inducing tissue. Notochord-derived Shh signals to the floor plate, and induces Shh expression in the floor plate. Floor plate-derived Shh subsequently signals to other cells in the neural tube, and is essential for proper specification of ventral neuron progenitor domains. Loss of Shh from the notochord and/or floor plate prevents proper specification of these progenitor domains. Shh binds Patched1, relieving Patched-mediated inhibition of Smoothened, leading to activation of the Gli family of transcription factors (GLI1, GLI2, and GLI3).

In this context Shh acts as a morphogen - it induces cell differentiation dependent on its concentration. At low concentrations it forms ventral interneurons, at higher concentrations it induces motor neuron development, and at highest concentrations it induces floor plate differentiation. Failure of Shh-modulated differentiation causes holoprosencephaly.

The dorsal neural tube is patterned by BMPs from the epidermal ectoderm flanking the neural plate. These induce sensory interneurons by activating Sr/Thr kinases and altering SMAD transcription factor levels.

Rostrocaudal (Anteroposterior) axis Edit

Signals that control anteroposterior neural development include FGF and retinoic acid, which act in the hindbrain and spinal cord.[15] The hindbrain, for example, is patterned by Hox genes, which are expressed in overlapping domains along the anteroposterior axis under the control of retinoic acid. The 3 (3 prime end) genes in the Hox cluster are induced by retinoic acid in the hindbrain, whereas the 5 (5 prime end) Hox genes are not induced by retinoic acid and are expressed more posteriorly in the spinal cord. Hoxb-1 is expressed in rhombomere 4 and gives rise to the facial nerve. Without this Hoxb-1 expression, a nerve similar to the trigeminal nerve arises.

Neurogenesis Edit

Neurogenesis is the process by which neurons are generated from neural stem cells and progenitor cells. Neurons are 'post-mitotic', meaning that they will never divide again for the lifetime of the organism.[9]

Epigenetic modifications play a key role in regulating gene expression in differentiating neural stem cells and are critical for cell fate determination in the developing and adult mammalian brain. Epigenetic modifications include DNA cytosine methylation to form 5-methylcytosine and 5-methylcytosine demethylation.[16][17] DNA cytosine methylation is catalyzed by DNA methyltransferases (DNMTs). Methylcytosine demethylation is catalyzed in several sequential steps by TET enzymes that carry out oxidative reactions (e.g. 5-methylcytosine to 5-hydroxymethylcytosine) and enzymes of the DNA base excision repair (BER) pathway.[16]

Neuronal migration Edit

 
Corticogenesis: younger neurons migrate past older ones using radial glia as a scaffolding. Cajal–Retzius cells (red) release reelin (orange).

Neuronal migration is the method by which neurons travel from their origin or birthplace to their final position in the brain. There are several ways they can do this, e.g. by radial migration or tangential migration. Sequences of radial migration (also known as glial guidance) and somal translocation have been captured by time-lapse microscopy.[18]

 
Tangential migration of interneurons from ganglionic eminence

Radial migration Edit

Neuronal precursor cells proliferate in the ventricular zone of the developing neocortex, where the principal neural stem cell is the radial glial cell. The first postmitotic cells must leave the stem cell niche and migrate outward to form the preplate, which is destined to become Cajal–Retzius cells and subplate neurons. These cells do so by somal translocation. Neurons migrating with this mode of locomotion are bipolar and attach the leading edge of the process to the pia. The soma is then transported to the pial surface by nucleokinesis, a process by which a microtubule "cage" around the nucleus elongates and contracts in association with the centrosome to guide the nucleus to its final destination.[19] Radial glial cells, whose fibers serve as a scaffolding for migrating cells and a means of radial communication mediated by calcium dynamic activity,[20][21] act as the main excitatory neuronal stem cell of the cerebral cortex[22][23] or translocate to the cortical plate and differentiate either into astrocytes or neurons.[24] Somal translocation can occur at any time during development.[18]

Subsequent waves of neurons split the preplate by migrating along radial glial fibres to form the cortical plate. Each wave of migrating cells travel past their predecessors forming layers in an inside-out manner, meaning that the youngest neurons are the closest to the surface.[25][26] It is estimated that glial guided migration represents 90% of migrating neurons in human and about 75% in rodents.[27]

Tangential migration Edit

Most interneurons migrate tangentially through multiple modes of migration to reach their appropriate location in the cortex. An example of tangential migration is the movement of interneurons from the ganglionic eminence to the cerebral cortex. One example of ongoing tangential migration in a mature organism, observed in some animals, is the rostral migratory stream connecting subventricular zone and olfactory bulb.

Axophilic migration Edit

Many neurons migrating along the anterior-posterior axis of the body use existing axon tracts to migrate along; this is called axophilic migration. An example of this mode of migration is in GnRH-expressing neurons, which make a long journey from their birthplace in the nose, through the forebrain, and into the hypothalamus.[28] Many of the mechanisms of this migration have been worked out, starting with the extracellular guidance cues[29] that trigger intracellular signaling. These intracellular signals, such as calcium signaling, lead to actin[30] and microtubule[31] cytoskeletal dynamics, which produce cellular forces that interact with the extracellular environment through cell adhesion proteins[32] to cause the movement of these cells.

Multipolar migration Edit

There is also a method of neuronal migration called multipolar migration.[33][34] This is seen in multipolar cells, which in the human, are abundantly present in the cortical intermediate zone. They do not resemble the cells migrating by locomotion or somal translocation. Instead these multipolar cells express neuronal markers and extend multiple thin processes in various directions independently of the radial glial fibers.[33]

Neurotrophic factors Edit

The survival of neurons is regulated by survival factors, called trophic factors. The neurotrophic hypothesis was formulated by Victor Hamburger and Rita Levi Montalcini based on studies of the developing nervous system. Victor Hamburger discovered that implanting an extra limb in the developing chick led to an increase in the number of spinal motor neurons. Initially he thought that the extra limb was inducing proliferation of motor neurons, but he and his colleagues later showed that there was a great deal of motor neuron death during normal development, and the extra limb prevented this cell death. According to the neurotrophic hypothesis, growing axons compete for limiting amounts of target-derived trophic factors and axons that fail to receive sufficient trophic support die by apoptosis. It is now clear that factors produced by a number of sources contribute to neuronal survival.

  • Nerve Growth Factor (NGF): Rita Levi Montalcini and Stanley Cohen purified the first trophic factor, Nerve Growth Factor (NGF), for which they received the Nobel Prize. There are three NGF-related trophic factors: BDNF, NT3, and NT4, which regulate survival of various neuronal populations. The Trk proteins act as receptors for NGF and related factors. Trk is a receptor tyrosine kinase. Trk dimerization and phosphorylation leads to activation of various intracellular signaling pathways including the MAP kinase, Akt, and PKC pathways.
  • CNTF: Ciliary neurotrophic factor is another protein that acts as a survival factor for motor neurons. CNTF acts via a receptor complex that includes CNTFRα, GP130, and LIFRβ. Activation of the receptor leads to phosphorylation and recruitment of the JAK kinase, which in turn phosphorylates LIFRβ. LIFRβ acts as a docking site for the STAT transcription factors. JAK kinase phosphorylates STAT proteins, which dissociate from the receptor and translocate to the nucleus to regulate gene expression.
  • GDNF: Glial derived neurotrophic factor is a member of the TGFb family of proteins, and is a potent trophic factor for striatal neurons. The functional receptor is a heterodimer, composed of type 1 and type 2 receptors. Activation of the type 1 receptor leads to phosphorylation of Smad proteins, which translocate to the nucleus to activate gene expression.

Synapse formation Edit

Neuromuscular junction Edit

Much of our understanding of synapse formation comes from studies at the neuromuscular junction. The transmitter at this synapse is acetylcholine. The acetylcholine receptor (AchR) is present at the surface of muscle cells before synapse formation. The arrival of the nerve induces clustering of the receptors at the synapse. McMahan and Sanes showed that the synaptogenic signal is concentrated at the basal lamina. They also showed that the synaptogenic signal is produced by the nerve, and they identified the factor as Agrin. Agrin induces clustering of AchRs on the muscle surface and synapse formation is disrupted in agrin knockout mice. Agrin transduces the signal via MuSK receptor to rapsyn. Fischbach and colleagues showed that receptor subunits are selectively transcribed from nuclei next to the synaptic site. This is mediated by neuregulins.

In the mature synapse each muscle fiber is innervated by one motor neuron. However, during development, many of the fibers are innervated by multiple axons. Lichtman and colleagues have studied the process of synapses elimination.[35] This is an activity-dependent event. Partial blockage of the receptor leads to retraction of corresponding presynaptic terminals. Later they used a connectomic approach, i.e., tracing out all the connections between motor neurons and muscle fibers, to characterize developmental synapse elimination on the level of a full circuit. Analysis confirmed the massive rewiring, 10-fold decrease in the number of synapses, that takes place as axons prune their motor units but add more synaptic areas at the NMJs with which they remain in contact.[36]

CNS synapses Edit

Agrin appears not to be a central mediator of CNS synapse formation and there is active interest in identifying signals that mediate CNS synaptogenesis. Neurons in culture develop synapses that are similar to those that form in vivo, suggesting that synaptogenic signals can function properly in vitro. CNS synaptogenesis studies have focused mainly on glutamatergic synapses. Imaging experiments show that dendrites are highly dynamic during development and often initiate contact with axons. This is followed by recruitment of postsynaptic proteins to the site of contact. Stephen Smith and colleagues have shown that contact initiated by dendritic filopodia can develop into synapses.

Induction of synapse formation by glial factors: Barres and colleagues made the observation that factors in glial conditioned media induce synapse formation in retinal ganglion cell cultures. Synapse formation in the CNS is correlated with astrocyte differentiation suggesting that astrocytes might provide a synaptogenic factor. The identity of the astrocytic factors is not yet known.

Neuroligins and SynCAM as synaptogenic signals: Sudhof, Serafini, Scheiffele and colleagues have shown that neuroligins and SynCAM can act as factors that induce presynaptic differentiation. Neuroligins are concentrated at the postsynaptic site and act via neurexins concentrated in the presynaptic axons. SynCAM is a cell adhesion molecule that is present in both pre- and post-synaptic membranes.

Activity dependent mechanisms in the assembly of neural circuits Edit

The processes of neuronal migration, differentiation and axon guidance are generally believed to be activity-independent mechanisms and rely on hard-wired genetic programs in the neurons themselves. Research findings however have implicated a role for activity-dependent mechanisms in mediating some aspects of these processes such as the rate of neuronal migration,[37] aspects of neuronal differentiation[38] and axon pathfinding.[39] Activity-dependent mechanisms influence neural circuit development and are crucial for laying out early connectivity maps and the continued refinement of synapses which occurs during development.[40] There are two distinct types of neural activity we observe in developing circuits -early spontaneous activity and sensory-evoked activity. Spontaneous activity occurs early during neural circuit development even when sensory input is absent and is observed in many systems such as the developing visual system,[41][42] auditory system,[43][44] motor system,[45] hippocampus,[46] cerebellum[47] and neocortex.[48]

Experimental techniques such as direct electrophysiological recording, fluorescence imaging using calcium indicators and optogenetic techniques have shed light on the nature and function of these early bursts of activity.[49][50] They have distinct spatial and temporal patterns during development[51] and their ablation during development has been known to result in deficits in network refinement in the visual system.[52] In the immature retina, waves of spontaneous action potentials arise from the retinal ganglion cells and sweep across the retinal surface in the first few postnatal weeks.[53] These waves are mediated by neurotransmitter acetylcholine in the initial phase and later on by glutamate.[54] They are thought to instruct the formation of two sensory maps- the retinotopic map and eye-specific segregation.[55] Retinotopic map refinement occurs in downstream visual targets in the brain-the superior colliculus (SC) and dorsal lateral geniculate nucleus (LGN).[56] Pharmacological disruption and mouse models lacking the β2 subunit of the nicotinic acetylcholine receptor has shown that the lack of spontaneous activity leads to marked defects in retinotopy and eye-specific segregation.[55]

Recent studies confirm that microglia, the resident immune cell of the brain, establish direct contacts with the cell bodies of developing neurons, and through these connections, regulate neurogenesis, migration, integration and the formation of neuronal networks in an activity-dependent manner.[57]

In the developing auditory system, developing cochlea generate bursts of activity which spreads across the inner hair cells and spiral ganglion neurons which relay auditory information to the brain.[58] ATP release from supporting cells triggers action potentials in inner hair cells.[59] In the auditory system, spontaneous activity is thought to be involved in tonotopic map formation by segregating cochlear neuron axons tuned to high and low frequencies.[58] In the motor system, periodic bursts of spontaneous activity are driven by excitatory GABA and glutamate during the early stages and by acetylcholine and glutamate at later stages.[60] In the developing zebrafish spinal cord, early spontaneous activity is required for the formation of increasingly synchronous alternating bursts between ipsilateral and contralateral regions of the spinal cord and for the integration of new cells into the circuit.[61] Motor neurons innervating the same twitch muscle fibers are thought to maintain synchronous activity which allows both neurons to remain in contact with the muscle fiber in adulthood.[36] In the cortex, early waves of activity have been observed in the cerebellum and cortical slices.[62] Once sensory stimulus becomes available, final fine-tuning of sensory-coding maps and circuit refinement begins to rely more and more on sensory-evoked activity as demonstrated by classic experiments about the effects of sensory deprivation during critical periods.[62]

Contemporary diffusion-weighted MRI techniques may also uncover the macroscopic process of axonal development. The connectome can be constructed from diffusion MRI data: the vertices of the graph correspond to anatomically labelled gray matter areas, and two such vertices, say u and v, are connected by an edge if the tractography phase of the data processing finds an axonal fiber that connects the two areas, corresponding to u and v.

Consensus Connectome Dynamics

Numerous braingraphs, computed from the Human Connectome Project can be downloaded from the http://braingraph.org site. The Consensus Connectome Dynamics (CCD) is a remarkable phenomenon that was discovered by continuously decreasing the minimum confidence-parameter at the graphical interface of the Budapest Reference Connectome Server.[63][64] The Budapest Reference Connectome Server (http://connectome.pitgroup.org) depicts the cerebral connections of n=418 subjects with a frequency-parameter k: For any k=1,2,...,n one can view the graph of the edges that are present in at least k connectomes. If parameter k is decreased one-by-one from k=n through k=1 then more and more edges appear in the graph, since the inclusion condition is relaxed. The surprising observation is that the appearance of the edges is far from random: it resembles a growing, complex structure, like a tree or a shrub (visualized on the animation on the left).

It is hypothesized in [65] that the growing structure copies the axonal development of the human brain: the earliest developing connections (axonal fibers) are common at most of the subjects, and the subsequently developing connections have larger and larger variance, because their variances are accumulated in the process of axonal development.

Synapse elimination Edit

Several motorneurons compete for each neuromuscular junction, but only one survives until adulthood.[35] Competition in vitro has been shown to involve a limited neurotrophic substance that is released, or that neural activity infers advantage to strong post-synaptic connections by giving resistance to a toxin also released upon nerve stimulation. In vivo, it is suggested that muscle fibres select the strongest neuron through a retrograde signal or that activity-dependent synapse elimination mechanisms determine the identity of the "winning" axon at a motor endplate.[36]

Mapping Edit

Brain mapping can show how an animal's brain changes throughout its lifetime. As of 2021, scientists mapped and compared the whole brains of eight C. elegans worms across their development on the neuronal level[66][67] and the complete wiring of a single mammalian muscle from birth to adulthood.[36]

Adult neurogenesis Edit

Neurogenesis also occurs in specific parts of the adult brain.

See also Edit

References Edit

  1. ^ . 2 November 2016. Archived from the original on 2016-11-02. Retrieved 1 May 2020.
  2. ^ "Neural Tube Defects". Retrieved 6 December 2011.
  3. ^ a b Gilbert, Scott (2006). Developmental biology (8th ed.). Sinauer Associates Publishers. pp. 373–379. ISBN 9780878932504.
  4. ^ Wolpert, Lewis (2015). Principles of development (Fifth ed.). Oxford University Press. ISBN 9780199678143. OCLC 914509705.
  5. ^ Wolpert 2015, pp. 522–526.
  6. ^ Saladin, Kenneth (2011). Anatomy & Physiology The Unity of Form and Function. New York: McGraw Hill. p. 514. ISBN 9780073378251.
  7. ^ a b Gato, A; Alonso, MI; Martín, C.; et al. (28 August 2014). "Embryonic cerebrospinal fluid in brain development: neural progenitor control". Croatian Medical Journal. 55 (4): 299–305. doi:10.3325/cmj.2014.55.299. PMC 4157377. PMID 25165044.
  8. ^ Gilbert, Scott (2013). Developmental Biology (Tenth ed.). Sinauer Associates Inc. ISBN 978-1605351926.[page needed]
  9. ^ a b Kandel, Eric R. (2006). Principles of neural science (5. ed.). Appleton and Lange: McGraw Hill. ISBN 978-0071390118.[page needed]
  10. ^ Croteau-Chonka, Elise C.; Dean, Douglas C., III; Remer, Justin; Dirks, Holly; O'Muircheartaigh, Jonathan; Deoni, Sean C.L. (15 October 2015). "Examining the relationships between cortical maturation and white matter myelination throughout early childhoold". NeuroImage. 125: 413–421. doi:10.1016/j.neuroimage.2015.10.038. PMC 4691410. PMID 26499814.{{cite journal}}: CS1 maint: multiple names: authors list (link)  
  11. ^ Wolpert 2015, pp. 163.
  12. ^ Estomih Mtui; Gregory Gruener (2006). Clinical Neuroanatomy and Neuroscience. Philadelphia: Saunders. p. 1. ISBN 978-1-4160-3445-2.
  13. ^ Chambers, S. M.; Fasano, C. A.; Papapetrou, E. P.; Tomishima, M.; Sadelain, M.; Studer, L. (2009). "Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling". Nature Biotechnology. 27 (3): 275–280. doi:10.1038/nbt.1529. PMC 2756723. PMID 19252484.
  14. ^ Jessell, Thomas M.; Kandel, Eric R.; Schwartz, James H. (2000). "Chapter 55". Principles of neural science (4th ed.). New York: McGraw-Hill. ISBN 978-0838577011.
  15. ^ Duester, G (September 2008). "Retinoic acid synthesis and signaling during early organogenesis". Cell. 134 (6): 921–31. doi:10.1016/j.cell.2008.09.002. PMC 2632951. PMID 18805086.
  16. ^ a b Wang, Zhiqin; Tang, Beisha; He, Yuquan; Jin, Peng (March 2016). "DNA methylation dynamics in neurogenesis". Epigenomics. 8 (3): 401–414. doi:10.2217/epi.15.119. PMC 4864063. PMID 26950681.
  17. ^ Noack, Florian; Pataskar, Abhijeet; Schneider, Martin; Buchholz, Frank; Tiwari, Vijay K; Calegari, Federico (27 February 2019). "Assessment and site-specific manipulation of DNA (hydroxy-)methylation during mouse corticogenesis". Life Science Alliance. 2 (2): e201900331. doi:10.26508/lsa.201900331. PMC 6394126. PMID 30814272.
  18. ^ a b Nadarajah B, Brunstrom J, Grutzendler J, Wong R, Pearlman A (2001). "Two modes of radial migration in early development of the cerebral cortex". Nat Neurosci. 4 (2): 143–50. doi:10.1038/83967. PMID 11175874. S2CID 6208462.
  19. ^ Samuels B, Tsai L (2004). "Nucleokinesis illuminated". Nat Neurosci. 7 (11): 1169–70. doi:10.1038/nn1104-1169. PMID 15508010. S2CID 11704754.
  20. ^ Rakic, P (May 1972). "Mode of cell migration to the superficial layers of fetal monkey neocortex". The Journal of Comparative Neurology. 145 (1): 61–83. doi:10.1002/cne.901450105. PMID 4624784. S2CID 41001390.
  21. ^ Rash, BG; Ackman, JB; Rakic, P (February 2016). "Bidirectional radial Ca(2+) activity regulates neurogenesis and migration during early cortical column formation". Science Advances. 2 (2): e1501733. Bibcode:2016SciA....2E1733R. doi:10.1126/sciadv.1501733. PMC 4771444. PMID 26933693.
  22. ^ Noctor, SC; Flint, AC; Weissman, TA; Dammerman, RS; Kriegstein, AR (8 February 2001). "Neurons derived from radial glial cells establish radial units in neocortex". Nature. 409 (6821): 714–20. Bibcode:2001Natur.409..714N. doi:10.1038/35055553. PMID 11217860. S2CID 3041502.
  23. ^ Tamamaki N, Nakamura K, Okamoto K, Kaneko T (September 2001). "Radial glia is a progenitor of neocortical neurons in the developing cerebral cortex". Neurosci. Res. 41 (1): 51–60. doi:10.1016/S0168-0102(01)00259-0. PMID 11535293. S2CID 2539488.
  24. ^ Miyata T, Kawaguchi A, Okano H, Ogawa M (September 2001). "Asymmetric inheritance of radial glial fibers by cortical neurons". Neuron. 31 (5): 727–41. doi:10.1016/S0896-6273(01)00420-2. PMID 11567613.
  25. ^ Nadarajah B, Parnavelas J (2002). "Modes of neuronal migration in the developing cerebral cortex". Nature Reviews Neuroscience. 3 (6): 423–32. doi:10.1038/nrn845. PMID 12042877. S2CID 38910547.
  26. ^ Rakic P (1972). "Mode of cell migration to the superficial layers of fetal monkey neocortex". Journal of Comparative Neurology. 145 (1): 61–83. doi:10.1002/cne.901450105. PMID 4624784. S2CID 41001390.
  27. ^ Letinic K, Zoncu R, Rakic P (June 2002). "Origin of GABAergic neurons in the human neocortex". Nature. 417 (6889): 645–9. Bibcode:2002Natur.417..645L. doi:10.1038/nature00779. PMID 12050665. S2CID 4349070.
  28. ^ Wray S (2010). "From nose to brain: development of gonadotrophin-releasing hormone-1 neurones". Journal of Neuroendocrinology. 22 (7): 743–753. doi:10.1111/j.1365-2826.2010.02034.x. PMC 2919238. PMID 20646175.
  29. ^ Giacobini P, Messina A, Wray S, Giampietro C, Crepaldi T, Carmeliet P, Fasolo A (2007). "Hepatocyte growth factor acts as a motogen and guidance signal for gonadotropin hormone-releasing hormone-1 neuronal migration" (PDF). Journal of Neuroscience. 27 (2): 431–445. doi:10.1523/JNEUROSCI.4979-06.2007. PMC 6672060. PMID 17215404.
  30. ^ Hutchins BI, Klenke U, Wray S (2013). "Calcium release-dependent actin flow in the leading process mediates axophilic migration". Journal of Neuroscience. 33 (28): 11361–71. doi:10.1523/JNEUROSCI.3758-12.2013. PMC 3724331. PMID 23843509.
  31. ^ Hutchins, B. Ian; Wray, Susan (2014). "Capture of microtubule plus-ends at the actin cortex promotes axophilic neuronal migration by enhancing microtubule tension in the leading process". Frontiers in Cellular Neuroscience. 8: 400. doi:10.3389/fncel.2014.00400. PMC 4245908. PMID 25505874.
  32. ^ Parkash J, Cimino I, Ferraris N, Casoni F, Wray S, Cappy H, Prevot V, Giacobini P (2012). "Suppression of β1-integrin in gonadotropin-releasing hormone cells disrupts migration and axonal extension resulting in severe reproductive alterations". Journal of Neuroscience. 32 (47): 16992–7002. doi:10.1523/JNEUROSCI.3057-12.2012. PMC 5238668. PMID 23175850.
  33. ^ a b Tabata H, Nakajima K (5 November 2003). "Multipolar migration: the third mode of radial neuronal migration in the developing cerebral cortex". Journal of Neuroscience. 23 (31): 9996–10001. doi:10.1523/JNEUROSCI.23-31-09996.2003. PMC 6740853. PMID 14602813.
  34. ^ Nadarajah B, Alifragis P, Wong R, Parnavelas J (2003). "Neuronal migration in the developing cerebral cortex: observations based on real-time imaging". Cereb Cortex. 13 (6): 607–11. doi:10.1093/cercor/13.6.607. PMID 12764035.
  35. ^ a b Turney, Stephen G.; Lichtman, Jeff W.; Harris, William A. (26 June 2012). "Reversing the Outcome of Synapse Elimination at Developing Neuromuscular Junctions In Vivo: Evidence for Synaptic Competition and Its Mechanism". PLOS Biology. 10 (6): e1001352. doi:10.1371/journal.pbio.1001352. PMC 3383738. PMID 22745601.
  36. ^ a b c d Meirovitch, Yaron; Kang, Kai; Draft, Ryan W.; Pavarino, Elisa C.; Henao E., Maria F.; Yang, Fuming; Turney, Stephen G.; Berger, Daniel R.; Peleg, Adi; Schalek, Richard L.; Lu, Ju L.; Tapia, Juan-Carlos; Lichtman, Jeff W. (September 2021). "Neuromuscular connectomes across development reveal synaptic ordering rules" (PDF). bioRxiv. doi:10.1101/2021.09.20.460480. S2CID 237598181.
  37. ^ Komuro, Hitoshi; Rakic, Pasko (August 1996). "Intracellular Ca2+ Fluctuations Modulate the Rate of Neuronal Migration". Neuron. 17 (2): 275–285. doi:10.1016/s0896-6273(00)80159-2. PMID 8780651.
  38. ^ Gu, X; Olson, E.C; Spitzer, N.C (1994). "Spontaneous neuronal calcium spikes and waves during early differentiation". Journal of Neuroscience. 14 (11): 6325–35. doi:10.1523/JNEUROSCI.14-11-06325.1994. PMC 6577261. PMID 7965039.
  39. ^ Hanson, M.G; Milner, L.D; Landmesser, L.T (2008). "Spontaneous early activity in the chick spinal cord influences distinct motor axon pathfinding decisions". Brain Res. Rev. 57 (1): 77–85. doi:10.1016/j.brainresrev.2007.06.021. PMC 2233604. PMID 17920131.
  40. ^ Kirkby, L.A; Sack, G.S; Firl, A; Feller, M.B (Dec 4, 2013). "A role for correlated spontaneous activity in the assembly of neural circuits". Neuron. 80 (5): 1129–44. doi:10.1016/j.neuron.2013.10.030. PMC 4560201. PMID 24314725.
  41. ^ Huberman, A.D. (2007). "Mechanisms of eye-specific visual circuit development". Current Opinion in Neurobiology. 17 (1): 73–80. doi:10.1016/j.conb.2007.01.005. PMID 17254766. S2CID 19418882.
  42. ^ Meister, M; Wong, R.O.L; Baylor, D.A; Shatz, C.J (1991). "Synchronous bursts of action potentials in ganglion cells of the developing retina". Science. 252 (5008): 939–43. Bibcode:1991Sci...252..939M. doi:10.1126/science.2035024. PMID 2035024.
  43. ^ Lippe, W.R (1994). "Rhythmic spontaneous activity in the developing avian auditory system". The Journal of Neuroscience. 14 (3): 1486–95. doi:10.1523/JNEUROSCI.14-03-01486.1994. PMC 6577532. PMID 8126550.
  44. ^ Jones, T.A; Jones, S.M; Paggett, K.C (15 October 2001). "Primordial rhythmic bursting in embryonic cochlear ganglion cells". The Journal of Neuroscience. 21 (20): 8129–35. doi:10.1523/JNEUROSCI.21-20-08129.2001. PMC 6763868. PMID 11588185.
  45. ^ O'Donovan, M.J (1999). "The origin of spontaneous activity in developing networks of the vertebrate nervous system". Current Opinion in Neurobiology. 9 (1): 94–104. doi:10.1016/s0959-4388(99)80012-9. PMID 10072366. S2CID 37387513.
  46. ^ Crepel, V; Aronov, D; Jorquera, I; Represa, A; Ben-Ari, Y; Cossart, R (2007). "A parturition-associated non synaptic coherent activity pattern in the developing hippocampus". Neuron. 54 (1): 105–120. doi:10.1016/j.neuron.2007.03.007. PMID 17408581.
  47. ^ Watt, A.J; Cuntz, H; Mori, M; Nusser, Z; Sjostrom, P.J; Hausser, M (2009). "Traveling waves in developing cerebellar cortex mediated by asymmetrical Purkinje cell connectivity". Nature Neuroscience. 12 (4): 463–73. doi:10.1038/nn.2285. PMC 2912499. PMID 19287389.
  48. ^ Corlew, Rebekah; Bosma, Martha M.; Moody, William J. (October 2004). "Spontaneous, synchronous electrical activity in neonatal mouse cortical neurones". The Journal of Physiology. 560 (2): 377–390. doi:10.1113/jphysiol.2004.071621. PMC 1665264. PMID 15297578.
  49. ^ Feller, Marla B. (April 1999). "Spontaneous Correlated Activity in Developing Neural Circuits". Neuron. 22 (4): 653–656. doi:10.1016/s0896-6273(00)80724-2. PMID 10230785.
  50. ^ O'Donovan, Michael J.; Chub, Nikolai; Wenner, Peter (October 1998). "Mechanisms of spontaneous activity in developing spinal networks". Journal of Neurobiology. 37 (1): 131–145. doi:10.1002/(sici)1097-4695(199810)37:1<131::aid-neu10>3.0.co;2-h. PMID 9777737.
  51. ^ Stafford, Ben K.; Sher, Alexander; Litke, Alan M.; Feldheim, David A. (October 2009). "Spatial-Temporal Patterns of Retinal Waves Underlying Activity-Dependent Refinement of Retinofugal Projections". Neuron. 64 (2): 200–212. doi:10.1016/j.neuron.2009.09.021. PMC 2771121. PMID 19874788.
  52. ^ Torborg, Christine L.; Feller, Marla B. (July 2005). "Spontaneous patterned retinal activity and the refinement of retinal projections". Progress in Neurobiology. 76 (4): 213–235. doi:10.1016/j.pneurobio.2005.09.002. PMID 16280194. S2CID 24563014.
  53. ^ Galli, L; Maffei, L (7 October 1988). "Spontaneous impulse activity of rat retinal ganglion cells in prenatal life". Science. 242 (4875): 90–91. Bibcode:1988Sci...242...90G. doi:10.1126/science.3175637. PMID 3175637.
  54. ^ Ford, Kevin J.; Feller, Marla B. (26 July 2011). "Assembly and disassembly of a retinal cholinergic network". Visual Neuroscience. 29 (1): 61–71. doi:10.1017/S0952523811000216. PMC 3982217. PMID 21787461.
  55. ^ a b Kirkby, Lowry A.; Sack, Georgeann S.; Firl, Alana; Feller, Marla B. (December 2013). "A Role for Correlated Spontaneous Activity in the Assembly of Neural Circuits". Neuron. 80 (5): 1129–1144. doi:10.1016/j.neuron.2013.10.030. PMC 4560201. PMID 24314725.
  56. ^ Ackman, James B.; Burbridge, Timothy J.; Crair, Michael C. (10 October 2012). "Retinal waves coordinate patterned activity throughout the developing visual system". Nature. 490 (7419): 219–225. Bibcode:2012Natur.490..219A. doi:10.1038/nature11529. PMC 3962269. PMID 23060192.
  57. ^ Cserép, Csaba; Schwarcz, Anett D.; Pósfai, Balázs; László, Zsófia I.; Kellermayer, Anna; Környei, Zsuzsanna; Kisfali, Máté; Nyerges, Miklós; Lele, Zsolt; Katona, István (September 2022). "Microglial control of neuronal development via somatic purinergic junctions". Cell Reports. 40 (12): 111369. doi:10.1016/j.celrep.2022.111369. PMC 9513806. PMID 36130488. S2CID 252416407.
  58. ^ a b Kandler, Karl; Clause, Amanda; Noh, Jihyun (10 May 2009). "Tonotopic reorganization of developing auditory brainstem circuits". Nature Neuroscience. 12 (6): 711–717. doi:10.1038/nn.2332. PMC 2780022. PMID 19471270.
  59. ^ Tritsch, Nicolas X; Rodríguez-Contreras, Adrián; Crins, Tom T H; Wang, Han Chin; Borst, J Gerard G; Bergles, Dwight E (1 August 2010). "Calcium action potentials in hair cells pattern auditory neuron activity before hearing onset". Nature Neuroscience. 13 (9): 1050–1052. doi:10.1038/nn.2604. PMC 2928883. PMID 20676105.
  60. ^ Momose-Sato, Yoko; Sato, Katsushige (2013). "Large-scale synchronized activity in the embryonic brainstem and spinal cord". Frontiers in Cellular Neuroscience. 7: 36. doi:10.3389/fncel.2013.00036. PMC 3625830. PMID 23596392.
  61. ^ Warp, Erica; Agarwal, Gautam; Wyart, Claire; Friedmann, Drew; Oldfield, Claire S.; Conner, Alden; Del Bene, Filippo; Arrenberg, Aristides B.; Baier, Herwig; Isacoff, Ehud Y. (January 2012). "Emergence of Patterned Activity in the Developing Zebrafish Spinal Cord". Current Biology. 22 (2): 93–102. doi:10.1016/j.cub.2011.12.002. PMC 3267884. PMID 22197243.
  62. ^ a b Sanes, Dan; Reh, Thomas; Harris, William (2012). Development of the Nervous System (Third ed.). Burlington MA: Elsevier. ISBN 9780123745392. OCLC 827948474.[page needed]
  63. ^ Szalkai, Balázs; Kerepesi, Csaba; Varga, Bálint; Grolmusz, Vince (May 2015). "The Budapest Reference Connectome Server v2.0". Neuroscience Letters. 595: 60–62. arXiv:1412.3151. doi:10.1016/j.neulet.2015.03.071. PMID 25862487. S2CID 6563189.
  64. ^ Szalkai, Balázs; Kerepesi, Csaba; Varga, Bálint; Grolmusz, Vince (15 September 2016). "Parameterizable consensus connectomes from the Human Connectome Project: the Budapest Reference Connectome Server v3.0". Cognitive Neurodynamics. 11 (1): 113–116. arXiv:1602.04776. doi:10.1007/s11571-016-9407-z. PMC 5264751. PMID 28174617.
  65. ^ Kerepesi, Csaba; Szalkai, Balázs; Varga, Bálint; Grolmusz, Vince; Shi, Yongtang (30 June 2016). "How to Direct the Edges of the Connectomes: Dynamics of the Consensus Connectomes and the Development of the Connections in the Human Brain". PLOS ONE. 11 (6): e0158680. arXiv:1509.05703. Bibcode:2016PLoSO..1158680K. doi:10.1371/journal.pone.0158680. PMC 4928947. PMID 27362431.
  66. ^ . phys.org. Douglas, Isle Of Man UK: Science X. Lunenfeld-Tanenbaum Research Institute. Archived from the original on 20 June 2022. Retrieved 21 September 2021.
  67. ^ Witvliet, Daniel; Mulcahy, Ben; Mitchell, James K.; Meirovitch, Yaron; Berger, Daniel R.; Wu, Yuelong; Liu, Yufang; Koh, Wan Xian; Parvathala, Rajeev; Holmyard, Douglas; Schalek, Richard L.; Shavit, Nir; Chisholm, Andrew D.; Lichtman, Jeff W.; Samuel, Aravinthan D. T.; Zhen, Mei (August 2021). "Connectomes across development reveal principles of brain maturation". Nature. 596 (7871): 257–261. Bibcode:2021Natur.596..257W. bioRxiv 10.1101/2020.04.30.066209v3. doi:10.1038/s41586-021-03778-8. ISSN 1476-4687. PMC 8756380. PMID 34349261. S2CID 236927815.

External links Edit

  • Neural Development (peer-reviewed open access journal).
  • Translating Neurodevelopmental Time Across Mammalian Species
  • The Child's Developing Brain
  • Brain Development
  • How poverty might change the brain
  • The Teenage Brain

development, nervous, system, this, article, about, neural, development, types, animals, including, humans, information, specific, human, nervous, system, humans, formation, stabilization, synapses, during, development, synaptic, stabilization, developmental, . This article is about neural development in all types of animals including humans For information specific to the human nervous system see Development of the nervous system in humans For formation and stabilization of synapses during development see Synaptic stabilization Developmental Neuroscience redirects here For the journal see Developmental Neuroscience journal Neural Development redirects here For the journal see Neural Development journal The development of the nervous system or neural development neurodevelopment refers to the processes that generate shape and reshape the nervous system of animals from the earliest stages of embryonic development to adulthood The field of neural development draws on both neuroscience and developmental biology to describe and provide insight into the cellular and molecular mechanisms by which complex nervous systems develop from nematodes and fruit flies to mammals Defects in neural development can lead to malformations such as holoprosencephaly and a wide variety of neurological disorders including limb paresis and paralysis balance and vision disorders and seizures 1 and in humans other disorders such as Rett syndrome Down syndrome and intellectual disability 2 Contents 1 Overview of vertebrate brain development 2 Aspects 3 Neural induction 4 Regionalization 5 Patterning of the nervous system 5 1 Dorsoventral axis 5 2 Rostrocaudal Anteroposterior axis 6 Neurogenesis 7 Neuronal migration 7 1 Radial migration 7 2 Tangential migration 7 3 Axophilic migration 7 4 Multipolar migration 8 Neurotrophic factors 9 Synapse formation 9 1 Neuromuscular junction 9 2 CNS synapses 9 3 Activity dependent mechanisms in the assembly of neural circuits 10 Synapse elimination 11 Mapping 12 Adult neurogenesis 13 See also 14 References 15 External linksOverview of vertebrate brain development Edit nbsp Diagram of the vertebrate nervous systemFurther information Development of the nervous system in humans The vertebrate central nervous system CNS is derived from the ectoderm the outermost germ layer of the embryo A part of the dorsal ectoderm becomes specified to neural ectoderm neuroectoderm that forms the neural plate along the dorsal side of the embryo 3 This is a part of the early patterning of the embryo including the invertebrate embryo that also establishes an anterior posterior axis 4 5 The neural plate is the source of the majority of neurons and glial cells of the CNS The neural groove forms along the long axis of the neural plate and the neural plate folds to give rise to the neural tube 6 When the tube is closed at both ends it is filled with embryonic cerebrospinal fluid 7 As the embryo develops the anterior part of the neural tube expands and forms three primary brain vesicles which become the forebrain prosencephalon midbrain mesencephalon and hindbrain rhombencephalon These simple early vesicles enlarge and further divide into the telencephalon future cerebral cortex and basal ganglia diencephalon future thalamus and hypothalamus mesencephalon future colliculi metencephalon future pons and cerebellum and myelencephalon future medulla 8 The CSF filled central chamber is continuous from the telencephalon to the central canal of the spinal cord and constitutes the developing ventricular system of the CNS Embryonic cerebrospinal fluid differs from that formed in later developmental stages and from adult CSF it influences the behavior of neural precursors 7 Because the neural tube gives rise to the brain and spinal cord any mutations at this stage in development can lead to fatal deformities like anencephaly or lifelong disabilities like spina bifida During this time the walls of the neural tube contain neural stem cells which drive brain growth as they divide many times Gradually some of the cells stop dividing and differentiate into neurons and glial cells which are the main cellular components of the CNS The newly generated neurons migrate to different parts of the developing brain to self organize into different brain structures Once the neurons have reached their regional positions they extend axons and dendrites which allow them to communicate with other neurons via synapses Synaptic communication between neurons leads to the establishment of functional neural circuits that mediate sensory and motor processing and underlie behavior 9 nbsp Flowchart of human brain developmentAspects EditSome landmarks of neural development include the birth and differentiation of neurons from stem cell precursors the migration of immature neurons from their birthplaces in the embryo to their final positions outgrowth of axons and dendrites from neurons guidance of the motile growth cone through the embryo towards postsynaptic partners the generation of synapses between these axons and their postsynaptic partners and finally the lifelong changes in synapses which are thought to underlie learning and memory Typically these neurodevelopmental processes can be broadly divided into two classes activity independent mechanisms and activity dependent mechanisms Activity independent mechanisms are generally believed to occur as hardwired processes determined by genetic programs played out within individual neurons These include differentiation migration and axon guidance to their initial target areas These processes are thought of as being independent of neural activity and sensory experience Once axons reach their target areas activity dependent mechanisms come into play Although synapse formation is an activity independent event modification of synapses and synapse elimination requires neural activity Developmental neuroscience uses a variety of animal models including the mouse Mus musculus the fruit fly Drosophila melanogaster the zebrafish Danio rerio the frog Xenopus laevis and the roundworm Caenorhabditis elegans Myelination formation of the lipid myelin sheath around neuronal axons is a process that is essential for normal brain function The myelin sheath provides insulation for the nerve impulse when communicating between neural systems Without it the impulse would be disrupted and the signal would not reach its target thus impairing normal functioning Because so much of brain development occurs in the prenatal stage and infancy it is crucial that myelination along with cortical development occur properly Magnetic resonance imaging MRI is a non invasive technique used to investigate myelination and cortical maturation the cortex is the outer layer of the brain composed of gray matter Rather than showing the actual myelin the MRI picks up on the myelin water fraction a measure of myelin content Multicomponent relaxometry MCR allow visualization and quantification of myelin content MCR is also useful for tracking white matter maturation which plays an important role in cognitive development It has been discovered that in infancy myelination occurs in a caudal cranial posterior to anterior pattern Because there is little evidence of a relationship between myelination and cortical thickness it was revealed that cortical thickness is independent of white matter This allows various aspects of the brain to grow simultaneously leading to a more fully developed brain 10 Neural induction EditDuring early embryonic development of the vertebrate the dorsal ectoderm becomes specified to give rise to the epidermis and the nervous system a part of the dorsal ectoderm becomes specified to neural ectoderm to form the neural plate which gives rise to the nervous system 3 11 The conversion of undifferentiated ectoderm to neuroectoderm requires signals from the mesoderm At the onset of gastrulation presumptive mesodermal cells move through the dorsal blastopore lip and form a layer of mesoderm in between the endoderm and the ectoderm Mesodermal cells migrate along the dorsal midline to give rise to the notochord that develops into the vertebral column Neuroectoderm overlying the notochord develops into the neural plate in response to a diffusible signal produced by the notochord The remainder of the ectoderm gives rise to the epidermis The ability of the mesoderm to convert the overlying ectoderm into neural tissue is called neural induction In the early embryo the neural plate folds outwards to form the neural groove Beginning in the future neck region the neural folds of this groove close to create the neural tube The formation of the neural tube from the ectoderm is called neurulation The ventral part of the neural tube is called the basal plate the dorsal part is called the alar plate The hollow interior is called the neural canal and the open ends of the neural tube called the neuropores close off 12 A transplanted blastopore lip can convert ectoderm into neural tissue and is said to have an inductive effect Neural inducers are molecules that can induce the expression of neural genes in ectoderm explants without inducing mesodermal genes as well Neural induction is often studied in Xenopus embryos since they have a simple body plan and there are good markers to distinguish between neural and non neural tissue Examples of neural inducers are the molecules noggin and chordin When embryonic ectodermal cells are cultured at low density in the absence of mesodermal cells they undergo neural differentiation express neural genes suggesting that neural differentiation is the default fate of ectodermal cells In explant cultures which allow direct cell cell interactions the same cells differentiate into epidermis This is due to the action of BMP4 a TGF b family protein that induces ectodermal cultures to differentiate into epidermis During neural induction noggin and chordin are produced by the dorsal mesoderm notochord and diffuse into the overlying ectoderm to inhibit the activity of BMP4 This inhibition of BMP4 causes the cells to differentiate into neural cells Inhibition of TGF b and BMP bone morphogenetic protein signaling can efficiently induce neural tissue from pluripotent stem cells 13 Regionalization EditIn a later stage of development the superior part of the neural tube flexes at the level of the future midbrain the mesencephalon at the mesencephalic flexure or cephalic flexure Above the mesencephalon is the prosencephalon future forebrain and beneath it is the rhombencephalon future hindbrain The alar plate of the prosencephalon expands to form the telencephalon which gives rise to the cerebral hemispheres whilst its basal plate becomes the diencephalon The optical vesicle which eventually become the optic nerve retina and iris forms at the basal plate of the prosencephalon Patterning of the nervous system EditIn chordates dorsal ectoderm forms all neural tissue and the nervous system Patterning occurs due to specific environmental conditions different concentrations of signaling molecules Dorsoventral axis Edit The ventral half of the neural plate is controlled by the notochord which acts as the organiser The dorsal half is controlled by the ectoderm plate which flanks either side of the neural plate 14 Ectoderm follows a default pathway to become neural tissue Evidence for this comes from single cultured cells of ectoderm which go on to form neural tissue This is postulated to be because of a lack of BMPs which are blocked by the organiser The organiser may produce molecules such as follistatin noggin and chordin that inhibit BMPs The ventral neural tube is patterned by sonic hedgehog Shh from the notochord which acts as the inducing tissue Notochord derived Shh signals to the floor plate and induces Shh expression in the floor plate Floor plate derived Shh subsequently signals to other cells in the neural tube and is essential for proper specification of ventral neuron progenitor domains Loss of Shh from the notochord and or floor plate prevents proper specification of these progenitor domains Shh binds Patched1 relieving Patched mediated inhibition of Smoothened leading to activation of the Gli family of transcription factors GLI1 GLI2 and GLI3 In this context Shh acts as a morphogen it induces cell differentiation dependent on its concentration At low concentrations it forms ventral interneurons at higher concentrations it induces motor neuron development and at highest concentrations it induces floor plate differentiation Failure of Shh modulated differentiation causes holoprosencephaly The dorsal neural tube is patterned by BMPs from the epidermal ectoderm flanking the neural plate These induce sensory interneurons by activating Sr Thr kinases and altering SMAD transcription factor levels Rostrocaudal Anteroposterior axis Edit Signals that control anteroposterior neural development include FGF and retinoic acid which act in the hindbrain and spinal cord 15 The hindbrain for example is patterned by Hox genes which are expressed in overlapping domains along the anteroposterior axis under the control of retinoic acid The 3 3 prime end genes in the Hox cluster are induced by retinoic acid in the hindbrain whereas the 5 5 prime end Hox genes are not induced by retinoic acid and are expressed more posteriorly in the spinal cord Hoxb 1 is expressed in rhombomere 4 and gives rise to the facial nerve Without this Hoxb 1 expression a nerve similar to the trigeminal nerve arises Neurogenesis EditNeurogenesis is the process by which neurons are generated from neural stem cells and progenitor cells Neurons are post mitotic meaning that they will never divide again for the lifetime of the organism 9 Epigenetic modifications play a key role in regulating gene expression in differentiating neural stem cells and are critical for cell fate determination in the developing and adult mammalian brain Epigenetic modifications include DNA cytosine methylation to form 5 methylcytosine and 5 methylcytosine demethylation 16 17 DNA cytosine methylation is catalyzed by DNA methyltransferases DNMTs Methylcytosine demethylation is catalyzed in several sequential steps by TET enzymes that carry out oxidative reactions e g 5 methylcytosine to 5 hydroxymethylcytosine and enzymes of the DNA base excision repair BER pathway 16 Neuronal migration Edit nbsp Corticogenesis younger neurons migrate past older ones using radial glia as a scaffolding Cajal Retzius cells red release reelin orange Neuronal migration is the method by which neurons travel from their origin or birthplace to their final position in the brain There are several ways they can do this e g by radial migration or tangential migration Sequences of radial migration also known as glial guidance and somal translocation have been captured by time lapse microscopy 18 nbsp Tangential migration of interneurons from ganglionic eminenceRadial migration Edit Neuronal precursor cells proliferate in the ventricular zone of the developing neocortex where the principal neural stem cell is the radial glial cell The first postmitotic cells must leave the stem cell niche and migrate outward to form the preplate which is destined to become Cajal Retzius cells and subplate neurons These cells do so by somal translocation Neurons migrating with this mode of locomotion are bipolar and attach the leading edge of the process to the pia The soma is then transported to the pial surface by nucleokinesis a process by which a microtubule cage around the nucleus elongates and contracts in association with the centrosome to guide the nucleus to its final destination 19 Radial glial cells whose fibers serve as a scaffolding for migrating cells and a means of radial communication mediated by calcium dynamic activity 20 21 act as the main excitatory neuronal stem cell of the cerebral cortex 22 23 or translocate to the cortical plate and differentiate either into astrocytes or neurons 24 Somal translocation can occur at any time during development 18 Subsequent waves of neurons split the preplate by migrating along radial glial fibres to form the cortical plate Each wave of migrating cells travel past their predecessors forming layers in an inside out manner meaning that the youngest neurons are the closest to the surface 25 26 It is estimated that glial guided migration represents 90 of migrating neurons in human and about 75 in rodents 27 Tangential migration Edit Most interneurons migrate tangentially through multiple modes of migration to reach their appropriate location in the cortex An example of tangential migration is the movement of interneurons from the ganglionic eminence to the cerebral cortex One example of ongoing tangential migration in a mature organism observed in some animals is the rostral migratory stream connecting subventricular zone and olfactory bulb Axophilic migration Edit Many neurons migrating along the anterior posterior axis of the body use existing axon tracts to migrate along this is called axophilic migration An example of this mode of migration is in GnRH expressing neurons which make a long journey from their birthplace in the nose through the forebrain and into the hypothalamus 28 Many of the mechanisms of this migration have been worked out starting with the extracellular guidance cues 29 that trigger intracellular signaling These intracellular signals such as calcium signaling lead to actin 30 and microtubule 31 cytoskeletal dynamics which produce cellular forces that interact with the extracellular environment through cell adhesion proteins 32 to cause the movement of these cells Multipolar migration Edit There is also a method of neuronal migration called multipolar migration 33 34 This is seen in multipolar cells which in the human are abundantly present in the cortical intermediate zone They do not resemble the cells migrating by locomotion or somal translocation Instead these multipolar cells express neuronal markers and extend multiple thin processes in various directions independently of the radial glial fibers 33 Neurotrophic factors EditThe survival of neurons is regulated by survival factors called trophic factors The neurotrophic hypothesis was formulated by Victor Hamburger and Rita Levi Montalcini based on studies of the developing nervous system Victor Hamburger discovered that implanting an extra limb in the developing chick led to an increase in the number of spinal motor neurons Initially he thought that the extra limb was inducing proliferation of motor neurons but he and his colleagues later showed that there was a great deal of motor neuron death during normal development and the extra limb prevented this cell death According to the neurotrophic hypothesis growing axons compete for limiting amounts of target derived trophic factors and axons that fail to receive sufficient trophic support die by apoptosis It is now clear that factors produced by a number of sources contribute to neuronal survival Nerve Growth Factor NGF Rita Levi Montalcini and Stanley Cohen purified the first trophic factor Nerve Growth Factor NGF for which they received the Nobel Prize There are three NGF related trophic factors BDNF NT3 and NT4 which regulate survival of various neuronal populations The Trk proteins act as receptors for NGF and related factors Trk is a receptor tyrosine kinase Trk dimerization and phosphorylation leads to activation of various intracellular signaling pathways including the MAP kinase Akt and PKC pathways CNTF Ciliary neurotrophic factor is another protein that acts as a survival factor for motor neurons CNTF acts via a receptor complex that includes CNTFRa GP130 and LIFRb Activation of the receptor leads to phosphorylation and recruitment of the JAK kinase which in turn phosphorylates LIFRb LIFRb acts as a docking site for the STAT transcription factors JAK kinase phosphorylates STAT proteins which dissociate from the receptor and translocate to the nucleus to regulate gene expression GDNF Glial derived neurotrophic factor is a member of the TGFb family of proteins and is a potent trophic factor for striatal neurons The functional receptor is a heterodimer composed of type 1 and type 2 receptors Activation of the type 1 receptor leads to phosphorylation of Smad proteins which translocate to the nucleus to activate gene expression Synapse formation EditNeuromuscular junction Edit Main article Neuromuscular junction Much of our understanding of synapse formation comes from studies at the neuromuscular junction The transmitter at this synapse is acetylcholine The acetylcholine receptor AchR is present at the surface of muscle cells before synapse formation The arrival of the nerve induces clustering of the receptors at the synapse McMahan and Sanes showed that the synaptogenic signal is concentrated at the basal lamina They also showed that the synaptogenic signal is produced by the nerve and they identified the factor as Agrin Agrin induces clustering of AchRs on the muscle surface and synapse formation is disrupted in agrin knockout mice Agrin transduces the signal via MuSK receptor to rapsyn Fischbach and colleagues showed that receptor subunits are selectively transcribed from nuclei next to the synaptic site This is mediated by neuregulins In the mature synapse each muscle fiber is innervated by one motor neuron However during development many of the fibers are innervated by multiple axons Lichtman and colleagues have studied the process of synapses elimination 35 This is an activity dependent event Partial blockage of the receptor leads to retraction of corresponding presynaptic terminals Later they used a connectomic approach i e tracing out all the connections between motor neurons and muscle fibers to characterize developmental synapse elimination on the level of a full circuit Analysis confirmed the massive rewiring 10 fold decrease in the number of synapses that takes place as axons prune their motor units but add more synaptic areas at the NMJs with which they remain in contact 36 CNS synapses Edit Agrin appears not to be a central mediator of CNS synapse formation and there is active interest in identifying signals that mediate CNS synaptogenesis Neurons in culture develop synapses that are similar to those that form in vivo suggesting that synaptogenic signals can function properly in vitro CNS synaptogenesis studies have focused mainly on glutamatergic synapses Imaging experiments show that dendrites are highly dynamic during development and often initiate contact with axons This is followed by recruitment of postsynaptic proteins to the site of contact Stephen Smith and colleagues have shown that contact initiated by dendritic filopodia can develop into synapses Induction of synapse formation by glial factors Barres and colleagues made the observation that factors in glial conditioned media induce synapse formation in retinal ganglion cell cultures Synapse formation in the CNS is correlated with astrocyte differentiation suggesting that astrocytes might provide a synaptogenic factor The identity of the astrocytic factors is not yet known Neuroligins and SynCAM as synaptogenic signals Sudhof Serafini Scheiffele and colleagues have shown that neuroligins and SynCAM can act as factors that induce presynaptic differentiation Neuroligins are concentrated at the postsynaptic site and act via neurexins concentrated in the presynaptic axons SynCAM is a cell adhesion molecule that is present in both pre and post synaptic membranes Activity dependent mechanisms in the assembly of neural circuits Edit Further information Activity dependent plasticity The processes of neuronal migration differentiation and axon guidance are generally believed to be activity independent mechanisms and rely on hard wired genetic programs in the neurons themselves Research findings however have implicated a role for activity dependent mechanisms in mediating some aspects of these processes such as the rate of neuronal migration 37 aspects of neuronal differentiation 38 and axon pathfinding 39 Activity dependent mechanisms influence neural circuit development and are crucial for laying out early connectivity maps and the continued refinement of synapses which occurs during development 40 There are two distinct types of neural activity we observe in developing circuits early spontaneous activity and sensory evoked activity Spontaneous activity occurs early during neural circuit development even when sensory input is absent and is observed in many systems such as the developing visual system 41 42 auditory system 43 44 motor system 45 hippocampus 46 cerebellum 47 and neocortex 48 Experimental techniques such as direct electrophysiological recording fluorescence imaging using calcium indicators and optogenetic techniques have shed light on the nature and function of these early bursts of activity 49 50 They have distinct spatial and temporal patterns during development 51 and their ablation during development has been known to result in deficits in network refinement in the visual system 52 In the immature retina waves of spontaneous action potentials arise from the retinal ganglion cells and sweep across the retinal surface in the first few postnatal weeks 53 These waves are mediated by neurotransmitter acetylcholine in the initial phase and later on by glutamate 54 They are thought to instruct the formation of two sensory maps the retinotopic map and eye specific segregation 55 Retinotopic map refinement occurs in downstream visual targets in the brain the superior colliculus SC and dorsal lateral geniculate nucleus LGN 56 Pharmacological disruption and mouse models lacking the b2 subunit of the nicotinic acetylcholine receptor has shown that the lack of spontaneous activity leads to marked defects in retinotopy and eye specific segregation 55 Recent studies confirm that microglia the resident immune cell of the brain establish direct contacts with the cell bodies of developing neurons and through these connections regulate neurogenesis migration integration and the formation of neuronal networks in an activity dependent manner 57 In the developing auditory system developing cochlea generate bursts of activity which spreads across the inner hair cells and spiral ganglion neurons which relay auditory information to the brain 58 ATP release from supporting cells triggers action potentials in inner hair cells 59 In the auditory system spontaneous activity is thought to be involved in tonotopic map formation by segregating cochlear neuron axons tuned to high and low frequencies 58 In the motor system periodic bursts of spontaneous activity are driven by excitatory GABA and glutamate during the early stages and by acetylcholine and glutamate at later stages 60 In the developing zebrafish spinal cord early spontaneous activity is required for the formation of increasingly synchronous alternating bursts between ipsilateral and contralateral regions of the spinal cord and for the integration of new cells into the circuit 61 Motor neurons innervating the same twitch muscle fibers are thought to maintain synchronous activity which allows both neurons to remain in contact with the muscle fiber in adulthood 36 In the cortex early waves of activity have been observed in the cerebellum and cortical slices 62 Once sensory stimulus becomes available final fine tuning of sensory coding maps and circuit refinement begins to rely more and more on sensory evoked activity as demonstrated by classic experiments about the effects of sensory deprivation during critical periods 62 Contemporary diffusion weighted MRI techniques may also uncover the macroscopic process of axonal development The connectome can be constructed from diffusion MRI data the vertices of the graph correspond to anatomically labelled gray matter areas and two such vertices say u and v are connected by an edge if the tractography phase of the data processing finds an axonal fiber that connects the two areas corresponding to u and v source source source source Consensus Connectome DynamicsNumerous braingraphs computed from the Human Connectome Project can be downloaded from the http braingraph org site The Consensus Connectome Dynamics CCD is a remarkable phenomenon that was discovered by continuously decreasing the minimum confidence parameter at the graphical interface of the Budapest Reference Connectome Server 63 64 The Budapest Reference Connectome Server http connectome pitgroup org depicts the cerebral connections of n 418 subjects with a frequency parameter k For any k 1 2 n one can view the graph of the edges that are present in at least k connectomes If parameter k is decreased one by one from k n through k 1 then more and more edges appear in the graph since the inclusion condition is relaxed The surprising observation is that the appearance of the edges is far from random it resembles a growing complex structure like a tree or a shrub visualized on the animation on the left It is hypothesized in 65 that the growing structure copies the axonal development of the human brain the earliest developing connections axonal fibers are common at most of the subjects and the subsequently developing connections have larger and larger variance because their variances are accumulated in the process of axonal development Synapse elimination EditMain article Synaptic pruning Several motorneurons compete for each neuromuscular junction but only one survives until adulthood 35 Competition in vitro has been shown to involve a limited neurotrophic substance that is released or that neural activity infers advantage to strong post synaptic connections by giving resistance to a toxin also released upon nerve stimulation In vivo it is suggested that muscle fibres select the strongest neuron through a retrograde signal or that activity dependent synapse elimination mechanisms determine the identity of the winning axon at a motor endplate 36 Mapping EditBrain mapping can show how an animal s brain changes throughout its lifetime As of 2021 scientists mapped and compared the whole brains of eight C elegans worms across their development on the neuronal level 66 67 and the complete wiring of a single mammalian muscle from birth to adulthood 36 Adult neurogenesis EditMain article Adult neurogenesis Neurogenesis also occurs in specific parts of the adult brain See also EditAxon guidance KCC2 Pioneer neuron Neural Darwinism Brain development timelines Malleable intelligence Role of cell adhesions in neural developmentReferences Edit Neurological Signs amp Diseases 2 November 2016 Archived from the original on 2016 11 02 Retrieved 1 May 2020 Neural Tube Defects Retrieved 6 December 2011 a b Gilbert Scott 2006 Developmental biology 8th ed Sinauer Associates Publishers pp 373 379 ISBN 9780878932504 Wolpert Lewis 2015 Principles of development Fifth ed Oxford University Press ISBN 9780199678143 OCLC 914509705 Wolpert 2015 pp 522 526 Saladin Kenneth 2011 Anatomy amp Physiology The Unity of Form and Function New York McGraw Hill p 514 ISBN 9780073378251 a b Gato A Alonso MI Martin C et al 28 August 2014 Embryonic cerebrospinal fluid in brain development neural progenitor control Croatian Medical Journal 55 4 299 305 doi 10 3325 cmj 2014 55 299 PMC 4157377 PMID 25165044 Gilbert Scott 2013 Developmental Biology Tenth ed Sinauer Associates Inc ISBN 978 1605351926 page needed a b Kandel Eric R 2006 Principles of neural science 5 ed Appleton and Lange McGraw Hill ISBN 978 0071390118 page needed Croteau Chonka Elise C Dean Douglas C III Remer Justin Dirks Holly O Muircheartaigh Jonathan Deoni Sean C L 15 October 2015 Examining the relationships between cortical maturation and white matter myelination throughout early childhoold NeuroImage 125 413 421 doi 10 1016 j neuroimage 2015 10 038 PMC 4691410 PMID 26499814 a href Template Cite journal html title Template Cite journal cite journal a CS1 maint multiple names authors list link nbsp Wolpert 2015 pp 163 Estomih Mtui Gregory Gruener 2006 Clinical Neuroanatomy and Neuroscience Philadelphia Saunders p 1 ISBN 978 1 4160 3445 2 Chambers S M Fasano C A Papapetrou E P Tomishima M Sadelain M Studer L 2009 Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling Nature Biotechnology 27 3 275 280 doi 10 1038 nbt 1529 PMC 2756723 PMID 19252484 Jessell Thomas M Kandel Eric R Schwartz James H 2000 Chapter 55 Principles of neural science 4th ed New York McGraw Hill ISBN 978 0838577011 Duester G September 2008 Retinoic acid synthesis and signaling during early organogenesis Cell 134 6 921 31 doi 10 1016 j cell 2008 09 002 PMC 2632951 PMID 18805086 a b Wang Zhiqin Tang Beisha He Yuquan Jin Peng March 2016 DNA methylation dynamics in neurogenesis Epigenomics 8 3 401 414 doi 10 2217 epi 15 119 PMC 4864063 PMID 26950681 Noack Florian Pataskar Abhijeet Schneider Martin Buchholz Frank Tiwari Vijay K Calegari Federico 27 February 2019 Assessment and site specific manipulation of DNA hydroxy methylation during mouse corticogenesis Life Science Alliance 2 2 e201900331 doi 10 26508 lsa 201900331 PMC 6394126 PMID 30814272 a b Nadarajah B Brunstrom J Grutzendler J Wong R Pearlman A 2001 Two modes of radial migration in early development of the cerebral cortex Nat Neurosci 4 2 143 50 doi 10 1038 83967 PMID 11175874 S2CID 6208462 Samuels B Tsai L 2004 Nucleokinesis illuminated Nat Neurosci 7 11 1169 70 doi 10 1038 nn1104 1169 PMID 15508010 S2CID 11704754 Rakic P May 1972 Mode of cell migration to the superficial layers of fetal monkey neocortex The Journal of Comparative Neurology 145 1 61 83 doi 10 1002 cne 901450105 PMID 4624784 S2CID 41001390 Rash BG Ackman JB Rakic P February 2016 Bidirectional radial Ca 2 activity regulates neurogenesis and migration during early cortical column formation Science Advances 2 2 e1501733 Bibcode 2016SciA 2E1733R doi 10 1126 sciadv 1501733 PMC 4771444 PMID 26933693 Noctor SC Flint AC Weissman TA Dammerman RS Kriegstein AR 8 February 2001 Neurons derived from radial glial cells establish radial units in neocortex Nature 409 6821 714 20 Bibcode 2001Natur 409 714N doi 10 1038 35055553 PMID 11217860 S2CID 3041502 Tamamaki N Nakamura K Okamoto K Kaneko T September 2001 Radial glia is a progenitor of neocortical neurons in the developing cerebral cortex Neurosci Res 41 1 51 60 doi 10 1016 S0168 0102 01 00259 0 PMID 11535293 S2CID 2539488 Miyata T Kawaguchi A Okano H Ogawa M September 2001 Asymmetric inheritance of radial glial fibers by cortical neurons Neuron 31 5 727 41 doi 10 1016 S0896 6273 01 00420 2 PMID 11567613 Nadarajah B Parnavelas J 2002 Modes of neuronal migration in the developing cerebral cortex Nature Reviews Neuroscience 3 6 423 32 doi 10 1038 nrn845 PMID 12042877 S2CID 38910547 Rakic P 1972 Mode of cell migration to the superficial layers of fetal monkey neocortex Journal of Comparative Neurology 145 1 61 83 doi 10 1002 cne 901450105 PMID 4624784 S2CID 41001390 Letinic K Zoncu R Rakic P June 2002 Origin of GABAergic neurons in the human neocortex Nature 417 6889 645 9 Bibcode 2002Natur 417 645L doi 10 1038 nature00779 PMID 12050665 S2CID 4349070 Wray S 2010 From nose to brain development of gonadotrophin releasing hormone 1 neurones Journal of Neuroendocrinology 22 7 743 753 doi 10 1111 j 1365 2826 2010 02034 x PMC 2919238 PMID 20646175 Giacobini P Messina A Wray S Giampietro C Crepaldi T Carmeliet P Fasolo A 2007 Hepatocyte growth factor acts as a motogen and guidance signal for gonadotropin hormone releasing hormone 1 neuronal migration PDF Journal of Neuroscience 27 2 431 445 doi 10 1523 JNEUROSCI 4979 06 2007 PMC 6672060 PMID 17215404 Hutchins BI Klenke U Wray S 2013 Calcium release dependent actin flow in the leading process mediates axophilic migration Journal of Neuroscience 33 28 11361 71 doi 10 1523 JNEUROSCI 3758 12 2013 PMC 3724331 PMID 23843509 Hutchins B Ian Wray Susan 2014 Capture of microtubule plus ends at the actin cortex promotes axophilic neuronal migration by enhancing microtubule tension in the leading process Frontiers in Cellular Neuroscience 8 400 doi 10 3389 fncel 2014 00400 PMC 4245908 PMID 25505874 Parkash J Cimino I Ferraris N Casoni F Wray S Cappy H Prevot V Giacobini P 2012 Suppression of b1 integrin in gonadotropin releasing hormone cells disrupts migration and axonal extension resulting in severe reproductive alterations Journal of Neuroscience 32 47 16992 7002 doi 10 1523 JNEUROSCI 3057 12 2012 PMC 5238668 PMID 23175850 a b Tabata H Nakajima K 5 November 2003 Multipolar migration the third mode of radial neuronal migration in the developing cerebral cortex Journal of Neuroscience 23 31 9996 10001 doi 10 1523 JNEUROSCI 23 31 09996 2003 PMC 6740853 PMID 14602813 Nadarajah B Alifragis P Wong R Parnavelas J 2003 Neuronal migration in the developing cerebral cortex observations based on real time imaging Cereb Cortex 13 6 607 11 doi 10 1093 cercor 13 6 607 PMID 12764035 a b Turney Stephen G Lichtman Jeff W Harris William A 26 June 2012 Reversing the Outcome of Synapse Elimination at Developing Neuromuscular Junctions In Vivo Evidence for Synaptic Competition and Its Mechanism PLOS Biology 10 6 e1001352 doi 10 1371 journal pbio 1001352 PMC 3383738 PMID 22745601 a b c d Meirovitch Yaron Kang Kai Draft Ryan W Pavarino Elisa C Henao E Maria F Yang Fuming Turney Stephen G Berger Daniel R Peleg Adi Schalek Richard L Lu Ju L Tapia Juan Carlos Lichtman Jeff W September 2021 Neuromuscular connectomes across development reveal synaptic ordering rules PDF bioRxiv doi 10 1101 2021 09 20 460480 S2CID 237598181 Komuro Hitoshi Rakic Pasko August 1996 Intracellular Ca2 Fluctuations Modulate the Rate of Neuronal Migration Neuron 17 2 275 285 doi 10 1016 s0896 6273 00 80159 2 PMID 8780651 Gu X Olson E C Spitzer N C 1994 Spontaneous neuronal calcium spikes and waves during early differentiation Journal of Neuroscience 14 11 6325 35 doi 10 1523 JNEUROSCI 14 11 06325 1994 PMC 6577261 PMID 7965039 Hanson M G Milner L D Landmesser L T 2008 Spontaneous early activity in the chick spinal cord influences distinct motor axon pathfinding decisions Brain Res Rev 57 1 77 85 doi 10 1016 j brainresrev 2007 06 021 PMC 2233604 PMID 17920131 Kirkby L A Sack G S Firl A Feller M B Dec 4 2013 A role for correlated spontaneous activity in the assembly of neural circuits Neuron 80 5 1129 44 doi 10 1016 j neuron 2013 10 030 PMC 4560201 PMID 24314725 Huberman A D 2007 Mechanisms of eye specific visual circuit development Current Opinion in Neurobiology 17 1 73 80 doi 10 1016 j conb 2007 01 005 PMID 17254766 S2CID 19418882 Meister M Wong R O L Baylor D A Shatz C J 1991 Synchronous bursts of action potentials in ganglion cells of the developing retina Science 252 5008 939 43 Bibcode 1991Sci 252 939M doi 10 1126 science 2035024 PMID 2035024 Lippe W R 1994 Rhythmic spontaneous activity in the developing avian auditory system The Journal of Neuroscience 14 3 1486 95 doi 10 1523 JNEUROSCI 14 03 01486 1994 PMC 6577532 PMID 8126550 Jones T A Jones S M Paggett K C 15 October 2001 Primordial rhythmic bursting in embryonic cochlear ganglion cells The Journal of Neuroscience 21 20 8129 35 doi 10 1523 JNEUROSCI 21 20 08129 2001 PMC 6763868 PMID 11588185 O Donovan M J 1999 The origin of spontaneous activity in developing networks of the vertebrate nervous system Current Opinion in Neurobiology 9 1 94 104 doi 10 1016 s0959 4388 99 80012 9 PMID 10072366 S2CID 37387513 Crepel V Aronov D Jorquera I Represa A Ben Ari Y Cossart R 2007 A parturition associated non synaptic coherent activity pattern in the developing hippocampus Neuron 54 1 105 120 doi 10 1016 j neuron 2007 03 007 PMID 17408581 Watt A J Cuntz H Mori M Nusser Z Sjostrom P J Hausser M 2009 Traveling waves in developing cerebellar cortex mediated by asymmetrical Purkinje cell connectivity Nature Neuroscience 12 4 463 73 doi 10 1038 nn 2285 PMC 2912499 PMID 19287389 Corlew Rebekah Bosma Martha M Moody William J October 2004 Spontaneous synchronous electrical activity in neonatal mouse cortical neurones The Journal of Physiology 560 2 377 390 doi 10 1113 jphysiol 2004 071621 PMC 1665264 PMID 15297578 Feller Marla B April 1999 Spontaneous Correlated Activity in Developing Neural Circuits Neuron 22 4 653 656 doi 10 1016 s0896 6273 00 80724 2 PMID 10230785 O Donovan Michael J Chub Nikolai Wenner Peter October 1998 Mechanisms of spontaneous activity in developing spinal networks Journal of Neurobiology 37 1 131 145 doi 10 1002 sici 1097 4695 199810 37 1 lt 131 aid neu10 gt 3 0 co 2 h PMID 9777737 Stafford Ben K Sher Alexander Litke Alan M Feldheim David A October 2009 Spatial Temporal Patterns of Retinal Waves Underlying Activity Dependent Refinement of Retinofugal Projections Neuron 64 2 200 212 doi 10 1016 j neuron 2009 09 021 PMC 2771121 PMID 19874788 Torborg Christine L Feller Marla B July 2005 Spontaneous patterned retinal activity and the refinement of retinal projections Progress in Neurobiology 76 4 213 235 doi 10 1016 j pneurobio 2005 09 002 PMID 16280194 S2CID 24563014 Galli L Maffei L 7 October 1988 Spontaneous impulse activity of rat retinal ganglion cells in prenatal life Science 242 4875 90 91 Bibcode 1988Sci 242 90G doi 10 1126 science 3175637 PMID 3175637 Ford Kevin J Feller Marla B 26 July 2011 Assembly and disassembly of a retinal cholinergic network Visual Neuroscience 29 1 61 71 doi 10 1017 S0952523811000216 PMC 3982217 PMID 21787461 a b Kirkby Lowry A Sack Georgeann S Firl Alana Feller Marla B December 2013 A Role for Correlated Spontaneous Activity in the Assembly of Neural Circuits Neuron 80 5 1129 1144 doi 10 1016 j neuron 2013 10 030 PMC 4560201 PMID 24314725 Ackman James B Burbridge Timothy J Crair Michael C 10 October 2012 Retinal waves coordinate patterned activity throughout the developing visual system Nature 490 7419 219 225 Bibcode 2012Natur 490 219A doi 10 1038 nature11529 PMC 3962269 PMID 23060192 Cserep Csaba Schwarcz Anett D Posfai Balazs Laszlo Zsofia I Kellermayer Anna Kornyei Zsuzsanna Kisfali Mate Nyerges Miklos Lele Zsolt Katona Istvan September 2022 Microglial control of neuronal development via somatic purinergic junctions Cell Reports 40 12 111369 doi 10 1016 j celrep 2022 111369 PMC 9513806 PMID 36130488 S2CID 252416407 a b Kandler Karl Clause Amanda Noh Jihyun 10 May 2009 Tonotopic reorganization of developing auditory brainstem circuits Nature Neuroscience 12 6 711 717 doi 10 1038 nn 2332 PMC 2780022 PMID 19471270 Tritsch Nicolas X Rodriguez Contreras Adrian Crins Tom T H Wang Han Chin Borst J Gerard G Bergles Dwight E 1 August 2010 Calcium action potentials in hair cells pattern auditory neuron activity before hearing onset Nature Neuroscience 13 9 1050 1052 doi 10 1038 nn 2604 PMC 2928883 PMID 20676105 Momose Sato Yoko Sato Katsushige 2013 Large scale synchronized activity in the embryonic brainstem and spinal cord Frontiers in Cellular Neuroscience 7 36 doi 10 3389 fncel 2013 00036 PMC 3625830 PMID 23596392 Warp Erica Agarwal Gautam Wyart Claire Friedmann Drew Oldfield Claire S Conner Alden Del Bene Filippo Arrenberg Aristides B Baier Herwig Isacoff Ehud Y January 2012 Emergence of Patterned Activity in the Developing Zebrafish Spinal Cord Current Biology 22 2 93 102 doi 10 1016 j cub 2011 12 002 PMC 3267884 PMID 22197243 a b Sanes Dan Reh Thomas Harris William 2012 Development of the Nervous System Third ed Burlington MA Elsevier ISBN 9780123745392 OCLC 827948474 page needed Szalkai Balazs Kerepesi Csaba Varga Balint Grolmusz Vince May 2015 The Budapest Reference Connectome Server v2 0 Neuroscience Letters 595 60 62 arXiv 1412 3151 doi 10 1016 j neulet 2015 03 071 PMID 25862487 S2CID 6563189 Szalkai Balazs Kerepesi Csaba Varga Balint Grolmusz Vince 15 September 2016 Parameterizable consensus connectomes from the Human Connectome Project the Budapest Reference Connectome Server v3 0 Cognitive Neurodynamics 11 1 113 116 arXiv 1602 04776 doi 10 1007 s11571 016 9407 z PMC 5264751 PMID 28174617 Kerepesi Csaba Szalkai Balazs Varga Balint Grolmusz Vince Shi Yongtang 30 June 2016 How to Direct the Edges of the Connectomes Dynamics of the Consensus Connectomes and the Development of the Connections in the Human Brain PLOS ONE 11 6 e0158680 arXiv 1509 05703 Bibcode 2016PLoSO 1158680K doi 10 1371 journal pone 0158680 PMC 4928947 PMID 27362431 Why a tiny worm s brain development could shed light on human thinking phys org Douglas Isle Of Man UK Science X Lunenfeld Tanenbaum Research Institute Archived from the original on 20 June 2022 Retrieved 21 September 2021 Witvliet Daniel Mulcahy Ben Mitchell James K Meirovitch Yaron Berger Daniel R Wu Yuelong Liu Yufang Koh Wan Xian Parvathala Rajeev Holmyard Douglas Schalek Richard L Shavit Nir Chisholm Andrew D Lichtman Jeff W Samuel Aravinthan D T Zhen Mei August 2021 Connectomes across development reveal principles of brain maturation Nature 596 7871 257 261 Bibcode 2021Natur 596 257W bioRxiv 10 1101 2020 04 30 066209v3 doi 10 1038 s41586 021 03778 8 ISSN 1476 4687 PMC 8756380 PMID 34349261 S2CID 236927815 External links EditNeural Development peer reviewed open access journal Translating Neurodevelopmental Time Across Mammalian Species The Child s Developing Brain Brain Development How poverty might change the brain The Teenage Brain Retrieved from https en wikipedia org w index php title Development of the nervous system amp oldid 1175437351 Neuronal migration, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.