Development of the nervous system

Last updated

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.

Contents

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

Diagram of the vertebrate nervous system NSdiagram.svg
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] [4] This is a part of the early patterning of the embryo (including the invertebrate embryo) that also establishes an anterior-posterior axis. [5] [6] 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. [7] When the tube is closed at both ends it is filled with embryonic cerebrospinal fluid. [8] 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). [9] 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. [8] 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 [10] . 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. [11]

Flowchart of human brain development Development of nervous system.svg
Flowchart of human brain development

Aspects

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. [12]

Neural induction

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] [13] 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. [14]

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. [15]

Regionalization

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

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

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. [16]

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

Signals that control anteroposterior neural development include FGF and retinoic acid, which act in the hindbrain and spinal cord. [17] 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

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. [11]

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. [18] [19] 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. [18]

Neuronal migration

Corticogenesis: younger neurons migrate past older ones using radial glia as a scaffolding. Cajal-Retzius cells (red) release reelin (orange). Corticogenesis in a wild-type mouse.png
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. [20]

Tangential migration of interneurons from ganglionic eminence Interneuron-radial glial interactions in the developing cerebral cortex.png
Tangential migration of interneurons from ganglionic eminence

Radial migration

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. [21] Radial glial cells, whose fibers serve as a scaffolding for migrating cells and a means of radial communication mediated by calcium dynamic activity, [22] [23] act as the main excitatory neuronal stem cell of the cerebral cortex [24] [25] or translocate to the cortical plate and differentiate either into astrocytes or neurons. [26] Somal translocation can occur at any time during development. [20]

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. [27] [28] It is estimated that glial guided migration represents 90% of migrating neurons in human and about 75% in rodents. [29]

Tangential migration

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

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. [30] Many of the mechanisms of this migration have been worked out, starting with the extracellular guidance cues [31] that trigger intracellular signaling. These intracellular signals, such as calcium signaling, lead to actin [32] and microtubule [33] cytoskeletal dynamics, which produce cellular forces that interact with the extracellular environment through cell adhesion proteins [34] to cause the movement of these cells.

Multipolar migration

There is also a method of neuronal migration called multipolar migration. [35] [36] 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. [35]

Neurotrophic factors

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.

Synapse formation

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. [37] 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. [38]

CNS synapses

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

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, [39] aspects of neuronal differentiation [40] and axon pathfinding. [41] 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. [42] 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, [43] [44] auditory system, [45] [46] motor system, [47] hippocampus, [48] cerebellum [49] and neocortex. [50]

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. [51] [52] They have distinct spatial and temporal patterns during development [53] and their ablation during development has been known to result in deficits in network refinement in the visual system. [54] 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. [55] These waves are mediated by neurotransmitter acetylcholine in the initial phase and later on by glutamate. [56] They are thought to instruct the formation of two sensory maps- the retinotopic map and eye-specific segregation. [57] Retinotopic map refinement occurs in downstream visual targets in the brain-the superior colliculus (SC) and dorsal lateral geniculate nucleus (LGN). [58] 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. [57]

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. [59]

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. [60] ATP release from supporting cells triggers action potentials in inner hair cells. [61] 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. [60] 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. [62] 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. [63] 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. [38] In the cortex, early waves of activity have been observed in the cerebellum and cortical slices. [64] 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. [64]

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. [65] [66] 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 [67] 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

Several motorneurons compete for each neuromuscular junction, but only one survives until adulthood. [37] 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. [38]

Mapping

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 [68] [69] and the complete wiring of a single mammalian muscle from birth to adulthood. [38]

Adult neurogenesis

Neurogenesis also occurs in specific parts of the adult brain.

See also

Related Research Articles

<span class="mw-page-title-main">Axon</span> Long projection on a neuron that conducts signals to other neurons

An axon, or nerve fiber, is a long, slender projection of a nerve cell, or neuron, in vertebrates, that typically conducts electrical impulses known as action potentials away from the nerve cell body. The function of the axon is to transmit information to different neurons, muscles, and glands. In certain sensory neurons, such as those for touch and warmth, the axons are called afferent nerve fibers and the electrical impulse travels along these from the periphery to the cell body and from the cell body to the spinal cord along another branch of the same axon. Axon dysfunction can be the cause of many inherited and acquired neurological disorders that affect both the peripheral and central neurons. Nerve fibers are classed into three types – group A nerve fibers, group B nerve fibers, and group C nerve fibers. Groups A and B are myelinated, and group C are unmyelinated. These groups include both sensory fibers and motor fibers. Another classification groups only the sensory fibers as Type I, Type II, Type III, and Type IV.

<span class="mw-page-title-main">Dendrite</span> Small projection on a neuron that receives signals

A dendrite or dendron is a branched protoplasmic extension of a nerve cell that propagates the electrochemical stimulation received from other neural cells to the cell body, or soma, of the neuron from which the dendrites project. Electrical stimulation is transmitted onto dendrites by upstream neurons via synapses which are located at various points throughout the dendritic tree.

<span class="mw-page-title-main">Neuron</span> Electrically excitable cell found in the nervous system of animals

Within a nervous system, a neuron, neurone, or nerve cell is an electrically excitable cell that fires electric signals called action potentials across a neural network. Neurons communicate with other cells via synapses, which are specialized connections that commonly use minute amounts of chemical neurotransmitters to pass the electric signal from the presynaptic neuron to the target cell through the synaptic gap.

<span class="mw-page-title-main">Nervous system</span> Part of an animal that coordinates actions and senses

In biology, the nervous system is the highly complex part of an animal that coordinates its actions and sensory information by transmitting signals to and from different parts of its body. The nervous system detects environmental changes that impact the body, then works in tandem with the endocrine system to respond to such events. Nervous tissue first arose in wormlike organisms about 550 to 600 million years ago. In vertebrates it consists of two main parts, the central nervous system (CNS) and the peripheral nervous system (PNS). The CNS consists of the brain and spinal cord. The PNS consists mainly of nerves, which are enclosed bundles of the long fibers or axons, that connect the CNS to every other part of the body. Nerves that transmit signals from the brain are called motor nerves or efferent nerves, while those nerves that transmit information from the body to the CNS are called sensory nerves or afferent. Spinal nerves are mixed nerves that serve both functions. The PNS is divided into three separate subsystems, the somatic, autonomic, and enteric nervous systems. Somatic nerves mediate voluntary movement. The autonomic nervous system is further subdivided into the sympathetic and the parasympathetic nervous systems. The sympathetic nervous system is activated in cases of emergencies to mobilize energy, while the parasympathetic nervous system is activated when organisms are in a relaxed state. The enteric nervous system functions to control the gastrointestinal system. Both autonomic and enteric nervous systems function involuntarily. Nerves that exit from the cranium are called cranial nerves while those exiting from the spinal cord are called spinal nerves.

<span class="mw-page-title-main">Cerebral cortex</span> Outer layer of the cerebrum of the mammalian brain

The cerebral cortex, also known as the cerebral mantle, is the outer layer of neural tissue of the cerebrum of the brain in humans and other mammals. The cerebral cortex mostly consists of the six-layered neocortex, with just 10% consisting of the allocortex. It is separated into two cortices, by the longitudinal fissure that divides the cerebrum into the left and right cerebral hemispheres. The two hemispheres are joined beneath the cortex by the corpus callosum. The cerebral cortex is the largest site of neural integration in the central nervous system. It plays a key role in attention, perception, awareness, thought, memory, language, and consciousness. The cerebral cortex is part of the brain responsible for cognition.

<span class="mw-page-title-main">Glia</span> Support cells in the nervous system

Glia, also called glial cells(gliocytes) or neuroglia, are non-neuronal cells in the central nervous system (brain and spinal cord) and the peripheral nervous system that do not produce electrical impulses. The neuroglia make up more than one half the volume of neural tissue in our body. They maintain homeostasis, form myelin in the peripheral nervous system, and provide support and protection for neurons. In the central nervous system, glial cells include oligodendrocytes, astrocytes, ependymal cells and microglia, and in the peripheral nervous system they include Schwann cells and satellite cells.

<span class="mw-page-title-main">Olfactory epithelium</span> Specialised epithelial tissue in the nasal cavity that detects odours

The olfactory epithelium is a specialized epithelial tissue inside the nasal cavity that is involved in smell. In humans, it measures 5 cm2 (0.78 sq in) and lies on the roof of the nasal cavity about 7 cm (2.8 in) above and behind the nostrils. The olfactory epithelium is the part of the olfactory system directly responsible for detecting odors.

<span class="mw-page-title-main">Astrocyte</span> Type of brain cell

Astrocytes, also known collectively as astroglia, are characteristic star-shaped glial cells in the brain and spinal cord. They perform many functions, including biochemical control of endothelial cells that form the blood–brain barrier, provision of nutrients to the nervous tissue, maintenance of extracellular ion balance, regulation of cerebral blood flow, and a role in the repair and scarring process of the brain and spinal cord following infection and traumatic injuries. The proportion of astrocytes in the brain is not well defined; depending on the counting technique used, studies have found that the astrocyte proportion varies by region and ranges from 20% to around 40% of all glia. Another study reports that astrocytes are the most numerous cell type in the brain. Astrocytes are the major source of cholesterol in the central nervous system. Apolipoprotein E transports cholesterol from astrocytes to neurons and other glial cells, regulating cell signaling in the brain. Astrocytes in humans are more than twenty times larger than in rodent brains, and make contact with more than ten times the number of synapses.

Synaptogenesis is the formation of synapses between neurons in the nervous system. Although it occurs throughout a healthy person's lifespan, an explosion of synapse formation occurs during early brain development, known as exuberant synaptogenesis. Synaptogenesis is particularly important during an individual's critical period, during which there is a certain degree of synaptic pruning due to competition for neural growth factors by neurons and synapses. Processes that are not used, or inhibited during their critical period will fail to develop normally later on in life.

Axon guidance is a subfield of neural development concerning the process by which neurons send out axons to reach their correct targets. Axons often follow very precise paths in the nervous system, and how they manage to find their way so accurately is an area of ongoing research.

<span class="mw-page-title-main">Rostral migratory stream</span> One path neural stem cells take to reach the olfactory bulb


The rostral migratory stream (RMS) is a specialized migratory route found in the brain of some animals along which neuronal precursors that originated in the subventricular zone (SVZ) of the brain migrate to reach the main olfactory bulb (OB). The importance of the RMS lies in its ability to refine and even change an animal's sensitivity to smells, which explains its importance and larger size in the rodent brain as compared to the human brain, as our olfactory sense is not as developed. This pathway has been studied in the rodent, rabbit, and both the squirrel monkey and rhesus monkey. When the neurons reach the OB they differentiate into GABAergic interneurons as they are integrated into either the granule cell layer or periglomerular layer.

A neurite or neuronal process refers to any projection from the cell body of a neuron. This projection can be either an axon or a dendrite. The term is frequently used when speaking of immature or developing neurons, especially of cells in culture, because it can be difficult to tell axons from dendrites before differentiation is complete.

<span class="mw-page-title-main">Radial glial cell</span> Bipolar-shaped progenitor cells of all neurons in the cerebral cortex and some glia

Radial glial cells, or radial glial progenitor cells (RGPs), are bipolar-shaped progenitor cells that are responsible for producing all of the neurons in the cerebral cortex. RGPs also produce certain lineages of glia, including astrocytes and oligodendrocytes. Their cell bodies (somata) reside in the embryonic ventricular zone, which lies next to the developing ventricular system.

<span class="mw-page-title-main">Synaptic pruning</span> Process of synapse elimination that occurs between early childhood and the onset of puberty

Synaptic pruning, a phase in the development of the nervous system, is the process of synapse elimination that occurs between early childhood and the onset of puberty in many mammals, including humans. Pruning starts near the time of birth and continues into the late-20s. During pruning, both the axon and dendrite decay and die off. It was traditionally considered to be complete by the time of sexual maturation, but this was discounted by MRI studies.

<span class="mw-page-title-main">Subgranular zone</span>

The subgranular zone (SGZ) is a brain region in the hippocampus where adult neurogenesis occurs. The other major site of adult neurogenesis is the subventricular zone (SVZ) in the brain.

The development of the nervous system in humans, or neural development or neurodevelopment involves the studies of embryology, developmental biology, and neuroscience to describe the cellular and molecular mechanisms by which the complex nervous system forms in humans, develops during prenatal development, and continues to develop postnatally.

Developmental plasticity is a general term referring to changes in neural connections during development as a result of environmental interactions as well as neural changes induced by learning. Much like neuroplasticity, or brain plasticity, developmental plasticity is specific to the change in neurons and synaptic connections as a consequence of developmental processes. A child creates most of these connections from birth to early childhood. There are three primary methods by which this may occur as the brain develops, but critical periods determine when lasting changes may form. Developmental plasticity may also be used in place of the term phenotypic plasticity when an organism in an embryonic or larval stage can alter its phenotype based on environmental factors. However, a main difference between the two is that phenotypic plasticity experienced during adulthood can be reversible, whereas traits that are considered developmentally plastic set foundations during early development that remain throughout the life of the organism.

Guidepost cells are cells which assist in the subcellular organization of both neural axon growth and migration. They act as intermediate targets for long and complex axonal growths by creating short and easy pathways, leading axon growth cones towards their target area.

The development of the cerebral cortex, known as corticogenesis is the process during which the cerebral cortex of the brain is formed as part of the development of the nervous system of mammals including its development in humans. The cortex is the outer layer of the brain and is composed of up to six layers. Neurons formed in the ventricular zone migrate to their final locations in one of the six layers of the cortex. The process occurs from embryonic day 10 to 17 in mice and between gestational weeks seven to 18 in humans.

<span class="mw-page-title-main">Brain cell</span> Functional tissue of the brain

Brain cells make up the functional tissue of the brain. The rest of the brain tissue is structural or connective called the stroma which includes blood vessels. The two main types of cells in the brain are neurons, also known as nerve cells, and glial cells, also known as neuroglia.

References

  1. "Neurological Signs & Diseases". 2 November 2016. Archived from the original on 2016-11-02. Retrieved 1 May 2020.
  2. "Neural Tube Defects" . Retrieved 6 December 2011.
  3. 1 2 Gilbert, Scott (2006). Developmental biology (8th ed.). Sinauer Associates Publishers. pp.  373–379. ISBN   978-0-87893-250-4.
  4. Zhou, Yi; Song, Hongjun; Ming, Guo-Li (2023-07-28). "Genetics of human brain development". Nature Reviews. Genetics. doi:10.1038/s41576-023-00626-5. ISSN   1471-0064. PMID   37507490.
  5. Wolpert, Lewis (2015). Principles of development (Fifth ed.). Oxford University Press. ISBN   978-0-19-967814-3. OCLC   914509705.
  6. Wolpert 2015, pp. 522–526.
  7. Saladin, Kenneth (2011). Anatomy & Physiology The Unity of Form and Function. New York: McGraw Hill. p. 514. ISBN   978-0-07-337825-1.
  8. 1 2 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.
  9. Gilbert, Scott (2013). Developmental Biology (Tenth ed.). Sinauer Associates Inc. ISBN   978-1-60535-192-6.[ page needed ]
  10. Zhou, Yi; Song, Hongjun; Ming, Guo-Li (2023-07-28). "Genetics of human brain development". Nature Reviews. Genetics. doi:10.1038/s41576-023-00626-5. ISSN   1471-0064. PMID   37507490.
  11. 1 2 Kandel, Eric R. (2006). Principles of neural science (5. ed.). Appleton and Lange: McGraw Hill. ISBN   978-0-07-139011-8.[ page needed ]
  12. 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. Open Access logo PLoS transparent.svg
  13. Wolpert 2015, pp. 163.
  14. Estomih Mtui; Gregory Gruener (2006). Clinical Neuroanatomy and Neuroscience. Philadelphia: Saunders. p. 1. ISBN   978-1-4160-3445-2.
  15. 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.
  16. Jessell, Thomas M.; Kandel, Eric R.; Schwartz, James H. (2000). "Chapter 55". Principles of neural science (4th ed.). New York: McGraw-Hill. ISBN   978-0-8385-7701-1.
  17. 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.
  18. 1 2 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.
  19. 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.
  20. 1 2 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.
  21. Samuels B, Tsai L (2004). "Nucleokinesis illuminated". Nat Neurosci. 7 (11): 1169–70. doi:10.1038/nn1104-1169. PMID   15508010. S2CID   11704754.
  22. 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.
  23. 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.
  24. 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.
  25. 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.
  26. 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.
  27. 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.
  28. 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.
  29. 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.
  30. 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.
  31. 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.
  32. 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.
  33. 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.
  34. 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.
  35. 1 2 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.
  36. 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.
  37. 1 2 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.
  38. 1 2 3 4 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.
  39. 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.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. 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.
  57. 1 2 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.
  58. 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.
  59. 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.
  60. 1 2 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.
  61. 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.
  62. 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.
  63. 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.
  64. 1 2 Sanes, Dan; Reh, Thomas; Harris, William (2012). Development of the Nervous System (Third ed.). Burlington MA: Elsevier. ISBN   978-0-12-374539-2. OCLC   827948474.[ page needed ]
  65. 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.
  66. 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.
  67. 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.
  68. "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.
  69. 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.