ElShamah - Reason & Science: Defending ID and the Christian Worldview
Would you like to react to this message? Create an account in a few clicks or log in to continue.
ElShamah - Reason & Science: Defending ID and the Christian Worldview

Otangelo Grasso: This is my library, where I collect information and present arguments developed by myself that lead, in my view, to the Christian faith, creationism, and Intelligent Design as the best explanation for the origin of the physical world.


You are not connected. Please login or register

Evolution of the brain

Go down  Message [Page 1 of 1]

1Evolution of the brain Empty Evolution of the brain Sat Aug 19, 2017 8:02 am

Otangelo


Admin

Evolution of the brain

http://reasonandscience.heavenforum.org/t2598-evolution-of-the-brain

The Platyhelminthes are located in an important position with respect to the evolution of metazoan . 3 They are widely recognized as being among the simplest organisms possessing three tissue layers (triploblasts), bilateral symmetry, cephalization, and complex organ systems. Planarians have characteristic organs along the anteroposterior axis, such as a pair of eyes and auricles, and a brain with simple architecture in the anterior head. One of the most notable characteristics of planarians is their high regenerative ability. They can regenerate whole animals, including a functional brain, from tiny fragments from almost any part of their bodies after amputation. An early worker on planarians described them as “almost immortal under the edge of the knife”. Later, Charles Darwin, famous as the author of The Origin of Species, was also interested in the regenerative ability of planarians. The robust regenerative abilities of planarians are based on a population of pluripotent stem cells called neoblasts, which are the only mitotic somatic cells in adults and are distributed throughout the body in planarians.

Evolution of the brain Planar10

Structural and Cellular Aspects of the Planarian Brain
In addition to planarian regenerative ability, they possess another important biological feature; that is, planarians belong to an evolutionarily early group that acquired a central nervous system (CNS). The planarian CNS is composed of two morphologically distinct structures: a bilobed brain, composed of about 2.0– 3.0 10^4 neurons in an adult planarian of about 8 mm in length, with nine branches on each outer side in the anterior region of the animal, and two longitudinal ventral nerve cords (VNCs) along the body (Fig. 4.1b). The brain is composed of a cortex of nerve cells in its outer region and a core of nerve fibers in its inner region (Fig. 4.2a). A pair of eyes is located on the dorsal side at the level of the third lateral branch of the brain. These morphological features of the brain structure suggested that external stimuli sensed by various organs, and the information thus acquired
by sensory neurons, might be accumulated inside the brain, and then processed and integrated to transduce the signals into the activity of motor neurons.

Flatworms are the earliest known animals to have a brain, and the simplest animals alive to have bilateral symmetry. They are also the simplest animals with organs that form from three germ layers. 1

THE URBILATERIAN BRAIN REVISITED: NOVEL INSIGHTS INTO OLD QUESTIONS FROM NEW FLATWORM CLADES 2
Flatworms are classically considered to represent the simplest organizational form of all living bilaterians with a true central nervous system. Based on their simple body plans, all flatworms have been traditionally grouped together in a single phylum at the base of the bilaterians. Current molecular phylogenomic studies now split the flatworms into two widely separated clades, the acoelomorph flatworms and the platyhelminth flatworms, such that the last common ancestor of both clades corresponds to the urbilaterian ancestor of all bilaterian animals. Remarkably, recent comparative neuroanatomical analyses of acoelomorphs and platyhelminths show that both of these flatworm groups have complex anterior brains with surprisingly similar basic neuroarchitectures. Taken together, these findings imply that fundamental neuroanatomical features of the brain in the two separate flatworm groups are likely to be primitive and derived from the urbilaterian brain.

CONSERVED DEVELOPMENTAL PROGRAMS FOR DIVERSE BILATERIAN BRAINS
At the structural level, the brains of higher deuterostomes such as vertebrates and higher protostomes such as arthropods or annelids are strikingly different. Moreover the embryological processes that give rise to these brains are also different in these two animal groups. The brain and dorsally located nerve cord of vertebrates derive from a dorsal neuroectoderm that invaginates to form a neural tube. In contrast, the brain and ventrally located ganglionic nerve cord of arthropods and annelids derive from a ventral neuroectoderm. In addition, the mechanism of neural progenitor proliferation shows significant differences. As shown for several arthropod taxa, but also probably true for other protostome phyla, asymmetrically dividing neural stem cells (neuroblasts) generate morphologically distinct lineages of neurons/glial cells which also form structural units of the brain. By contrast, neural progenitors in vertebrates form a layer of symmetrically dividing cells. These cells eventually switch to asymmetric divisions when producing neurons, but no evidence exists to date that neurons descending from individual progenitors form structural units, such as individual brainstem nuclei, or cortical layers. These and other differences in CNS structure and development have been used as one basis for the classification of “vertebrate-like” notoneuralia versus “invertebrate-like” gastroneuralia types. It is interesting to ask what the CNS of the common bilaterian ancestor looked like, and how one can envisage the evolutionary changes that led to the divergence of the two types of nervous systems.

Key developmental processes such as regionalization of the neural primordium and specification of certain cell types are conserved in brain development of protostomes and deuterostomes. This implies that the brains of all bilaterian animals are evolutionarily related and derive from an ancestral urbilaterian ancestor which may have already possessed a developmental genetic program for brain architecture of considerable complexity.

Classically, flatworms are considered to have the simplest organization of all bilaterians which is characterized by a lack of coelom, respiratory system, circulatory system, skeletal system, and through-gut. Flatworms have been traditionally grouped together in a single phylum, the members of which are thought to have been least changed from the ancestral bilaterian form. Accordingly, many traditional phylogenies placed this classical platyhelminth monophylum in a group of “acoelomates” at a basal position in the bilaterian tree. Given this phylogenetic perspective, the flatworm brain might also be least changed from the ancestral form and hence most representative of the urbilaterian brain. This notion is in accordance with classical comparative neuroanatomical studies that considered flatworms to be the most primitive bilaterians possessing a true central nervous system.

With a look towards earlier evolutionary stages, i.e., to the medusa or polyp-like ancestor of flatworms, it was suggested that the orthogon derives from the diffuse nerve net whereby concentrations of neurons at certain places (e.g., around the mouth or tentacles, coalesce into coherent fiber tracts; looking forward towards “higher animals”, it was hypothesized that a further concentration and restriction of neural elements had ensued, in such a way that in the ancestors of protostomes the dorsal tracts of the orthogon were eliminated, and in the ancestors of chordates the ventral ones.

Evolution of the brain Flatwo10

With the advent of molecular phylogenetics a decade ago, a major revision of the classical bilaterian phylogeny became necessary. In this revision, the entire platyhelminth phylum was removed from its basal position and firmly embedded within the lophotrochozoans, one of the two protostome superclades (Fig. 2B). As a result, the platyhelminth flatworms could no longer be considered to be more basal than any of the other lophotrochozoan phyla such as the annelids or molluscs. From this revised phylogenetic point of view there is no a priori reason to assume that the flatworm CNS should manifest primitive features characteristic of the urbilaterian brain. Indeed based on this revised phylogeny none of the living animals would correspond to intermediates between protostomes and deuterostomes, hence, making it more difficult to reconstruct the anatomical organization of the ancestral bilaterian brain from any extant species.

Recent neuroanatomical studies demonstrate that both types of flatworms have relatively complex anterior brains that consist of a cortex of neural cell bodies and a central neuropile with numerous commissural and longitudinal fiber bundles. Early classical histological analyses of the central nervous systems of acoelomorph flatworms reported the presence of a bilobed central brain composed of numerous neuronal cell bodies associated with complex commissural and connective fiber bundles


1. https://en.wikipedia.org/wiki/Timeline_of_human_evolution#Hominidae
2. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3873165/
3. Brain Evolution by Design, Shuichi Shigeno Yasunori Murakami Tadashi Nomura, page 82

https://reasonandscience.catsboard.com

2Evolution of the brain Empty Evolution of the human brain Tue Aug 22, 2017 4:56 pm

Otangelo


Admin

Evolution of the human brain

Evolution of the brain 53e33810

The human brain, due to evolution, or design ?!
Brain Evolution Ralph L. Holloway
Department of Anthropology, Columbia University, New York, NY,
SYNOPSIS
The size of the hominid brain increased from about 450 ml at 3.5 million years ago to our current average volume of 1350 ml. These changes through time were sometimes gradual but not always.
Now let's make a little calculation. The human brain has 1500000000000000 synapses. According to above claim, the hominid brain of our ur-ancestor, 3,5mio years ago, had a brain, a third of the size of homo sapiens today, that is 500000000000000 synapses approximately. That means there was an increase in a number of brain synapses of 1000000000000000 in 3.500.000 years. That means, there had to be an increase of 1.000.000.000.000.000: 3.500.000 of approximately 285700000 per year, or 782000 per day, or 32600 per hour.
In computing terms, the brain’s nerve cells, called neurons, are the processors, while synapses, the junctions where neurons meet and transmit information to each other, are analogous to memory. These synapses are not " just so" interconnected. The connections process and store information and must be the correct ones..... like a computer network.

Just for comparison:

The brain is a deviously complex biological computing device that even the fastest supercomputers in the world fail to emulate. 3 Well, that’s not entirely true anymore. Researchers at the Okinawa Institute of Technology Graduate University in Japan and Forschungszentrum Jülich in Germany have managed to simulate a single second of human brain activity in a very, very powerful computer. It took 40 minutes with the combined muscle of 82,944 processors in K computer to get just 1 second of biological brain processing time.
So how could natural selection, genetic drift or gene flow have produced the correct 32600 brain connections average per hour during 3,5bio years?
Sounds legit.....

12/30/2004  1
The cover story in Cell1 this week has set off a flurry of startling headlines: EurekAlert pronounces, “Evidence that human brain evolution was a special event” and “University of Chicago researchers discovered that humans are a ‘privileged’ evolutionary lineage.”The gist of the research by Dorus et al. from the Howard Hughes Medical Institute and University of Chicago is that there is a huge genetic gap between human brains and those of our nearest alleged ancestors.  EurekAlert explains:

One of the study’s major surprises is the relatively large number of genes that have contributed to human brain evolution.  “For a long time, people have debated about the genetic underpinning of human brain evolution,” said [Bruce] Lahn [HHMI}.  “Is it a few mutations in a few genes, a lot of mutations in a few genes, or a lot of mutations in a lot of genes?  The answer appears to be a lot of mutations in a lot of genes.  We’ve done a rough calculation that the evolution of the human brain probably involves hundreds if not thousands of mutations in perhaps hundreds or thousands of genes — and even that is a conservative estimate.”It is nothing short spectacular that so many mutations in so many genes were acquired during the mere 20-25 million years of time in the evolutionary lineage leading to humans, according to Lahn.  This means that selection has worked “extra-hard” during human evolution to create the powerful brain that exists in humans.

Doruset al., “Accelerated Evolution of Nervous System Genes in the Origin of Homo sapiens,” Cell Volume 119, Issue 7, 29 December 2004, Pages 1027-1040, doi:10.1016/j.cell.2004.11.040.

The science outlets are spinning this story without letting go of Darwinism.  They are throwing around phrases like strong selection, intensified selection and other nonsense as if random mutations conspired to sculpt the most complex piece of matter in the known universe.  They know better.  Orthogenesis (straight-line evolution) is out.  Teleology is out.  Personifying natural selection is out, so all they have to work with are thousands of random, undirected changes over thousands of different genes that have no ability to conspire with one another.  (In fact, they counteract one another see 11/29/2004 and 10/19/2004 headlines).  But if even one beneficial mutation is hard to find (see 03/19/2002 headline), how is any rational person to believe that thousands – “and that is a conservative estimate” – accomplished such a feat?  The gig is up, Darwin Party: surrender.  It’s over.  Throw down your arms.  The award for Stupid Evolution Quote of the Week goes to Bruce Lahn for his one-liner that “selection has worked ‘extra-hard’ during human evolution to create the powerful brain that exists in humans.”  This can serve as USO entertainment for the liberation troops as they begin their clean-up operations.

Brain Evolution
Ralph L. Holloway
Department of Anthropology, Columbia University, New York, NY, USA
SYNOPSIS
The evolution of the human brain has been a combination of reorganization of brain components, and increases of brain size through both hyperplasia and hypertrophy during development, underlain by neurogenomic changes. Paleoneurology based on endocast studies is the direct evidence demonstrating volume changes through time, and if present, some convolutional details of the underlying cerebral cortex. Reorganizational changes include a reduction of primary visual cortex and relative enlargement of posterior association cortex and expanded Broca’s regions, as well as cerebral asymmetries. The size of the hominid brain increased from about 450 ml at 3.5 million years ago to our current average volume of 1350 ml. These changes through time were sometimes gradual but not always.

Now let's make a little calculation. The human brain has 1500000000000000 synapses. According to above claim, the hominid brain of our ur-ancestor, 3,5mio years ago, had a brain, a third of the size of homo sapiens today, that is 500000000000000 synapses approximately. That means there was an increase in a number of brain synapses of 1000000000000000 in 3.500.000 years.  That means, there had to be an increase of 1.000.000.000.000.000: 3.500.000 of approximately 285.700.000  per year, or  782.000 per day, or 32.600 per hour.

Now let's suppose the average age of each generation was 50 years. that means that there would have been 70000 generations in 3,5mio years. That means, in each generation, there would have had to be an increase of 14300000000, or 14,3 billion new synapse connections per generation.....

1. http://creationsafaris.com/crev200412.htm#earlyman116
2. Basics in Human Evolution, Michael P. Muehlenbein,  page 232
3. https://www.extremetech.com/extreme/163051-simulating-1-second-of-human-brain-activity-takes-82944-processors

https://reasonandscience.catsboard.com

3Evolution of the brain Empty Re: Evolution of the brain Wed Aug 23, 2017 6:34 pm

Otangelo


Admin

Evolution of the brain Brain_10
Evolution of the brain Brain_13
Evolution of the brain Brain_12
Evolution of the brain Brain_11
Evolution of the brain Brain_14

https://reasonandscience.catsboard.com

4Evolution of the brain Empty Re: Evolution of the brain Tue Sep 05, 2017 1:17 pm

Otangelo


Admin

Evolution of the brain Brain_15

https://reasonandscience.catsboard.com

5Evolution of the brain Empty Neural Crest Cells Migration Fri Sep 01, 2023 4:00 am

Otangelo


Admin

Neural Crest Cells Migration

Neural Crest Cells: A Central Player in Vertebrate Development

Neural crest cells (NCCs) are a transient, multipotent population of cells that arise at the border of the developing neural tube in vertebrate embryos. They are unique to vertebrates and have been termed the "fourth germ layer" due to their essential roles in development, despite not being a traditional embryonic layer like ectoderm, mesoderm, or endoderm.

Origin and Migration

Formation: Neural crest cells emerge during the process of neurulation. As the neural plate folds to form the neural tube, the cells at its dorsal edge (or crest) are specified as neural crest cells.
Migration: After their specification, NCCs undergo an epithelial-to-mesenchymal transition (EMT). This transition allows them to delaminate from the neural tube and migrate to various regions throughout the developing embryo.

Differentiation and Derivatives

NCCs are renowned for their remarkable pluripotency. Depending on their axial level of origin (cranial, trunk, vagal, etc.) and their environmental cues, they can differentiate into a wide array of cell types:

Cranial NCCs: Contribute to facial cartilage and bones, certain cranial ganglia, and connective tissues in the head.
Cardiac NCCs: Participate in the formation of the outflow tract of the heart.
Trunk NCCs: Differentiate into dorsal root ganglia (sensory ganglia of the spinal cord), sympathetic ganglia, adrenal medulla, and melanocytes.
Enteric NCCs: Colonize the entire gut to form the enteric nervous system.
Vagal and Sacral NCCs: Contribute to the enteric nervous system of the gut.

Significance in Development and Evolution

NCCs play pivotal roles in organ and tissue development, but they also have evolutionary significance. The emergence of NCCs in vertebrates has been associated with the evolution of novel vertebrate features, like the intricately structured face and skull, and the complex peripheral nervous system. Furthermore, the adaptive capabilities of NCCs, such as generating pigmented cells for protective coloration or forming structures for predatory or defensive strategies, have been crucial in vertebrate evolution.

Neural Crest Cells Migration

Neural crest migration is a crucial developmental event that underscores the intricacy and specificity of cellular movement during embryogenesis. These cells, arising from the neural crest region, give rise to a myriad of cell types and contribute to various structures, solidifying their importance in shaping organismal form and function. Here's an overview:

Neural Crest Cells (NCCs)

Origination: NCCs emerge from the dorsal neural tube during neurulation. Following their formation, they undergo an epithelial-to-mesenchymal transition (EMT), enabling their migration.
Migration Routes: Depending on their anteroposterior and dorsoventral origin, NCCs follow specific pathways. Cranial NCCs migrate to the pharyngeal arches, while trunk NCCs can follow either a dorsolateral path (beneath the ectoderm) or a ventromedial path (between the somite and neural tube).
Differentiation: Post-migration, NCCs differentiate into a multitude of cell types, including neurons, glial cells, melanocytes, cartilage and bone of the facial skeleton, and more.
Guidance Mechanisms: NCCs rely on a plethora of signaling molecules, receptors, and extracellular matrix components to ensure their precise migration. They interpret environmental cues to decide their direction, speed, and final destination.

Significance in Development

Diverse Derivatives: NCCs contribute to the formation of diverse tissues and structures, underscoring their indispensable role in embryonic development.
Facial Morphology: Cranial NCCs are pivotal in forming the facial cartilage and bones, playing a central role in shaping facial morphology.
Peripheral Nervous System: The peripheral nervous system owes its existence to NCCs, as they differentiate into sensory neurons, sympathetic and parasympathetic neurons, and Schwann cells.
Melanocytes: The melanocytes, responsible for skin and hair pigmentation, are products of the neural crest lineage.
Heart Development: Cardiac NCCs influence the septation of the outflow tract of the heart, ensuring the proper division of the aorta and pulmonary artery.

How do neural crest cells migrate and differentiate into various cell types, contributing to diverse structures?

Neural crest cells (NCCs) are remarkable for their migratory abilities and potential to give rise to a vast array of cell types. Their journey from the dorsal region of the neural tube to various parts of the embryo is both intricate and meticulously regulated.

Migration of Neural Crest Cells

Initiation of Migration: For NCCs to migrate, they first undergo an epithelial-to-mesenchymal transition (EMT). This process involves a loss of cell adhesion properties and acquisition of a motile cell phenotype, enabling them to delaminate from the neural tube.

Pathways

Depending on their axial origin, NCCs follow specific migratory pathways.  Cranial NCCs migrate in streams to populate the pharyngeal arches and form facial structures. Trunk NCCs take either a dorsolateral path between the ectoderm and somites, giving rise to melanocytes, or a ventromedial path between somites and the neural tube, differentiating into neurons and glia of the peripheral nervous system. Vagal and sacral NCCs migrate to the gut to form the enteric nervous system.

Guidance Mechanisms: NCC migration is guided by a combination of repulsive and attractive cues. These include: Extracellular matrix components, like fibronectin, that guide cells. Chemotactic molecules, like semaphorins or ephrins, that either attract or repel NCCs. Gap junction communications between migrating NCCs.

Differentiation of Neural Crest Cells

Once NCCs reach their destinations, they differentiate into specific cell types based on local environmental cues:

Cranial NCCs: They can become bones, cartilage, tendons, and connective tissues of the face and neck. They also contribute to cranial ganglia.
Trunk NCCs: These cells can become:

Melanocytes, responsible for pigmentation.
Sensory neurons and glial cells in the dorsal root ganglia.
Adrenal medulla cells.
Sympathetic chain ganglia.
Vagal and Sacral NCCs: These primarily differentiate into neurons and glia of the enteric nervous system.

Cardiac NCCs: They contribute to the septation of the cardiac outflow tract.

Regulation of Differentiation

Differentiation is regulated by a combination of intrinsic transcriptional programs and extrinsic signals from surrounding tissues. Key factors include:

Bone morphogenetic proteins (BMPs)
Wnt signaling
Sox proteins
Notch signaling

Clinical Significance

Understanding the migratory and differentiation patterns of NCCs is crucial for grasping the etiology of several congenital disorders, termed neurocristopathies. For instance, disruptions in NCC migration can lead to conditions like Hirschsprung's disease or DiGeorge syndrome.

What molecular cues guide neural crest cell migration and destination determination?

Neural crest cell (NCC) migration is a highly orchestrated process, requiring precise spatiotemporal regulation. Various molecular cues, both repulsive and attractive, ensure that NCCs follow specific paths and reach their intended destinations. Here are some of the prominent molecular players in this ballet:

Extracellular Matrix (ECM) Components

Fibronectin: NCCs preferentially migrate along fibronectin-rich paths. This adhesive protein helps in the initial detachment of NCCs from the neural tube and guides their migration.
Laminin: Found in the basement membranes, laminin interacts with integrin receptors on NCCs, aiding in migration.

Ephrins and Eph Receptors

Ephrin ligands and their Eph receptors control the directionality of NCC migration. For example:
EphB: expressed in the mesoderm, creates a repulsion mechanism preventing NCCs from entering this region.
EphrinB: guides the migration of cranial NCCs into the pharyngeal arches.

Semaphorins and Neuropilins/Plexins

Semaphorins: A family of secreted and membrane-bound proteins known to repel NCCs away from particular regions. Sema3A, for example, creates a barrier around the neural tube, directing NCCs into specific migratory streams.

Chemokine Signaling

Chemokines are small proteins that guide cell migration:

CXCL12/CXCR4: This chemokine/receptor pair is crucial for guiding cardiac neural crest cells to the outflow tract of the heart.

Wnt Signaling

Wnts: are secreted proteins that play roles in various cellular processes, including NCC migration. Wnt proteins can either attract or repel NCCs based on context.

Bone Morphogenetic Proteins (BMPs)

BMP2 and BMP4: are involved in the delamination and onset of migration of NCCs. They also play roles in determining the fate of migrating NCCs.

Notch Signaling

The Notch pathway is involved in maintaining the balance between cell proliferation and differentiation:

Dll1: a Notch ligand, is critical for the segregation and boundary formation between NCCs and the neural tube.

Slit/Robo Signaling

Slit proteins: These are repulsive cues that help guide NCC migration. For example, Slit2 can prevent NCCs from entering specific areas, ensuring precise migration paths.

The migration and final localization of neural crest cells are directed by an intricate interplay of molecular cues that help ensure the cells traverse the correct paths and differentiate appropriately. Disruptions in these molecular signals can lead to various developmental anomalies, underscoring their critical importance in embryonic development.

How do neural crest cell migration mechanisms contribute to the diversity of vertebrate structures?

Neural crest cells are remarkable for their multipotency and migratory capacity. Originating at the border of the neural plate and ectoderm, these cells embark on intricate migratory routes throughout the developing embryo, differentiating into a myriad of cell types and contributing to an impressive array of tissues and structures. Their versatility and broad range of derivatives underscore their pivotal role in vertebrate evolution and the complexity of vertebrate structures.

Migration Mechanisms of Neural Crest Cells:

Epithelial-to-Mesenchymal Transition (EMT): Before migration, neural crest cells undergo EMT, a process wherein they lose their epithelial characteristics, such as cell-to-cell adhesion, and acquire mesenchymal properties, including increased motility.
Guidance Cues: As they migrate, neural crest cells interpret a variety of molecular signals in their environment, such as chemotactic factors, which guide their movement and influence their ultimate destinations.
Extracellular Matrix (ECM) Interaction: Neural crest cells utilize their filopodia (thin cellular projections) to probe and navigate the ECM, adhering to and moving along its fibers.

Contributions to Vertebrate Structural Diversity:

Craniofacial Structures: Neural crest cells contribute to the cartilage, bone, and connective tissues of the face and anterior skull. The adaptability and diversity of these structures across vertebrates, from the beaks of birds to the jaws of mammals, can be attributed to the differentiation potential of neural crest cells.
Peripheral Nervous System (PNS): Neural crest cells give rise to the entire PNS, which includes sensory ganglia, sympathetic and parasympathetic chains, and Schwann cells. The evolution and diversification of the PNS have been instrumental in the sensory and motor adaptabilities of vertebrates.
Pigment Cells: Melanocytes, responsible for skin, hair, and eye coloration in vertebrates, are derived from neural crest cells. The wide array of pigmentation patterns and adaptations seen across vertebrate species can be traced back to these cells.
Cardiovascular Adaptations: In the heart and great vessels, neural crest cells contribute to the septation of the outflow tract, ensuring the separate circulation of oxygenated and deoxygenated blood, a feature crucial for the evolutionary success of warm-blooded vertebrates.
Endocrine and Other Cells: Neural crest cells also differentiate into endocrine cells like those of the adrenal medulla and even some connective tissue cells, adding to the myriad ways they influence vertebrate physiology and form.

The migratory and differentiation capabilities of neural crest cells have been instrumental in the evolutionary diversification of vertebrate structures. Their contributions span a wide range of tissues and systems, underscoring their pivotal role in vertebrate development and adaptability.

Appearance of Neural Crest Migration in the Evolutionary Timeline

Neural Crest Migration in the Evolutionary Timeline

The neural crest is a group of cells that emerge from the dorsal aspect of the neural tube during embryonic development. These cells migrate to various parts of the embryo and differentiate into a wide variety of cell types, playing crucial roles in the development of various tissues and organs. Understanding the appearance of neural crest migration provides insights into the intricacies of vertebrate evolution.

Origin of Neural Crest Cells


The appearance of neural crest cells marks a significant event in vertebrate evolution. It is hypothesized that these cells first appeared in early chordates, providing them with the ability to form complex structures such as cranial nerves and cartilage.

Migration Pathways:

Dorsal Pathway: After emerging from the neural tube, some neural crest cells would have taken a dorsal route, contributing to the formation of melanocytes in the skin and hair.
Ventral Pathway: Neural crest cells migrating through the ventral pathway would have contributed to the formation of neurons and glia of the peripheral nervous system.

Contribution to Craniofacial Structures

One of the most significant contributions of neural crest cells is in the formation of craniofacial structures. These cells would have migrated into the pharyngeal arches, giving rise to parts of the face, jaw, and throat in vertebrates.

Development of Peripheral Nervous System

Neural crest cells would have been integral in the development of the peripheral nervous system. They would have differentiated into sensory neurons, sympathetic and parasympathetic neurons, and Schwann cells.

Heart and Vascular Development

Neural crest cells also play a role in cardiovascular development. They would have contributed to the formation of the outflow tract of the heart and the aortic arches.

Evolutionary Significance

The appearance of neural crest cells and their migratory abilities would have provided early vertebrates with a distinct evolutionary advantage. The ability of these cells to differentiate into a wide range of cell types would have paved the way for the development of more complex structures and functions, setting the stage for the diverse array of vertebrates seen today.

De Novo Genetic Information Necessary to Instantiate Neural Crest Migration

Neural crest cells are multipotent migratory cells that originate from the dorsal neural tube in vertebrates. Their migration and subsequent differentiation are essential for the formation of diverse cell types and structures. The genetic orchestration underpinning the emergence, migration, and differentiation of neural crest cells is intricate. Here are some key genetic elements and processes that would be crucial for the initiation and execution of neural crest migration:

Neural Crest Induction

Neural Plate Border Specification: During early embryogenesis, signals such as BMP, Wnt, and FGF establish the neural plate border, which is the precursor to the neural crest.
Neural Crest Specifiers: Genes like Snail, Slug, FoxD3, and Sox10 are critical for determining the neural crest cell fate. Their expression marks the onset of neural crest development.

Migration of Neural Crest Cells

E-cadherin Downregulation: The neural crest cells undergo an epithelial-to-mesenchymal transition (EMT), enabling them to migrate. Downregulation of E-cadherin, a cell adhesion molecule, is a pivotal step in this transition.
Expression of N-cadherin and Neural Cell Adhesion Molecule (NCAM): Post EMT, the expression of N-cadherin and NCAM facilitates the migratory ability of neural crest cells.
Guidance Molecules: Several molecules like ephrins and semaphorins guide the migration paths of neural crest cells, ensuring they reach their target destinations.

Differentiation and Integration

Cardiac Neural Crest: These cells contribute to the formation of the outflow tract in the heart. Tbx1 and Nkx2.5 are essential genes associated with cardiac neural crest differentiation.
Trunk Neural Crest: Responsible for melanocytes and peripheral neurons. Key genes include Mitf and Ednrb.
Neural Crest Stem Cells (NCSCs): Genes like p75NTR and Sox10 characterize these cells, which retain the potential to differentiate into various cell types even after the embryonic period.

Manufacturing Codes and Languages Employed for Neural Crest Migration

Genetic Codes (Transcriptional Regulation)

Induction and Specification Genes: Genes such as Snail, Slug, FoxD3, Sox9, and Sox10 are involved in the early stages of neural crest cell induction and specification.
EMT Transition: The epithelial-to-mesenchymal transition (EMT) is crucial for neural crest cells to acquire migratory abilities. Key genes involved include Snail and Slug, which downregulate E-cadherin expression, facilitating EMT.

Signaling Pathways (Molecular Languages)

BMP, Wnt, and FGF Pathways: These signaling pathways are involved in the initial specification of neural plate border cells, which eventually give rise to neural crest cells.
Ephrin-Eph Signaling: This pathway provides guidance cues to migrating neural crest cells, ensuring that they follow specific paths during their journey.
Notch Signaling: Critical in maintaining the balance between cell proliferation and differentiation among neural crest cell populations.

Cell Adhesion Codes

Cadherins: As neural crest cells undergo EMT, there's a switch from E-cadherin to N-cadherin, promoting their migratory phenotype.
Integrins: These are cell adhesion molecules that help neural crest cells attach to and migrate along specific substrates.

Morphogen Gradients (Spatial Codes)

Chemokine Signaling: Chemokines and their receptors help create gradients that attract or repel migrating neural crest cells, ensuring they reach their intended destinations.

Post-Translational Modifications (Regulatory Codes)

Ubiquitination and Phosphorylation: These modifications can rapidly alter protein function, affecting neural crest cell behavior, including migration and differentiation.

Non-Coding RNA Language

microRNAs: These small RNA molecules can post-transcriptionally regulate gene expression, and several of them have been implicated in neural crest development and migration.

In essence, these "codes" and "languages" constitute a highly coordinated and regulated network of interactions and pathways that dictate the proper development of neural crest cells, ensuring they reach their correct destinations and differentiate appropriately.

Epigenetic Regulatory Mechanisms for Neural Crest Migration

The term "epigenetics" refers to modifications in gene expression that don't involve changes to the underlying DNA sequence. These modifications can be influenced by various factors like age, environment, and disease state. Epigenetic mechanisms play a pivotal role in neural crest cell formation, migration, and differentiation. Here are the primary epigenetic regulatory mechanisms implicated in neural crest migration:

DNA Methylation

Role in Neural Crest: DNA methylation involves the addition of a methyl group to the cytosine base in DNA. It's generally associated with gene repression. In the context of the neural crest, dynamic changes in methylation patterns are crucial for the induction and subsequent migration of these cells. For example, genes crucial for neural crest specification might be demethylated (and thus activated) at specific developmental stages.

Histone Modifications

Histone Acetylation and Deacetylation: The addition or removal of acetyl groups on histones can either promote or repress gene transcription. Histone deacetylases (HDACs) are known to influence neural crest migratory behavior, with HDAC inhibitors being able to modulate neural crest migration in developmental models.
Histone Methylation: Depending on the specific lysine residue that's modified on the histone, methylation can either activate or repress gene expression. Dynamic histone methylation events are critical for various stages of neural crest development, from induction to differentiation.

Non-Coding RNAs

microRNAs (miRNAs): These short RNA sequences can bind to mRNA and either degrade them or prevent their translation, thereby influencing gene expression. Specific miRNAs are known to be crucial for neural crest formation, EMT, migration, and differentiation.
Long Non-Coding RNAs (lncRNAs): While less is known about their specific roles in neural crest cells compared to miRNAs, lncRNAs have been shown to play roles in various developmental processes, including those of the neural crest.

Chromatin Remodeling

Role in Neural Crest: Chromatin remodeling complexes can shift, evict, or restructure nucleosomes, thereby making DNA more or less accessible for transcription. The SWI/SNF chromatin remodeling complex, for instance, is known to play a role in neural crest development.

RNA Methylation

N6-methyladenosine (m6A) Modification: Recent studies have identified m6A RNA modifications in influencing neural crest development, showcasing the dynamic and multifaceted epigenetic controls in place.

Neural crest migration is orchestrated by a symphony of genetic and epigenetic cues. Understanding these epigenetic regulatory mechanisms not only sheds light on normal developmental processes but can also offer insights into developmental disorders where neural crest cell function is disrupted.

Signaling Pathways for Neural Crest Migration

Neural crest cells (NCCs) are a group of multipotent cells that originate from the dorsal neural tube and undergo migration to contribute to various cell lineages and tissues in vertebrates. The migration of neural crest cells is a tightly regulated process, orchestrated by a multitude of signaling pathways that ensure proper spatial and temporal patterns of cell movement. Below are the key signaling pathways involved in guiding neural crest migration:

Bone Morphogenetic Protein (BMP) Signaling

Role in Neural Crest Migration: BMPs play a fundamental role in establishing the neural plate border and inducing neural crest cell formation. BMP signaling influences the expression of neural crest specifiers like Snail, Slug, and FoxD3.

Wnt Signaling

Role in Neural Crest Migration: Canonical and non-canonical Wnt pathways are instrumental for neural crest induction, EMT, and migration. Wnt signaling promotes the expression of neural crest markers and plays a role in determining the directionality of neural crest cell migration.

Fibroblast Growth Factor (FGF) Signaling

Role in Neural Crest Migration: FGF signaling is involved in the early stages of neural crest induction and also influences the migratory capabilities of NCCs.

Ephrin-Eph Signaling

Role in Neural Crest Migration: Eph receptors and their ephrin ligands act as repulsive cues guiding migrating neural crest cells. This signaling helps establish distinct migratory streams and prevents the mixing of neural crest cell populations.

Notch Signaling

Role in Neural Crest Migration: Notch signaling contributes to neural crest lineage decisions and also plays a role in modulating cell migration.

Retinoic Acid Signaling

Role in Neural Crest Migration: Retinoic acid gradients help define the anterior-posterior axis in the embryo and play a critical role in cranial neural crest migration and patterning.

Chemokine Signaling

Role in Neural Crest Migration: Chemokines like CXCL12 and its receptor CXCR4 have been identified as guides for neural crest migration, directing the cells towards regions of high ligand concentration.

Platelet-derived Growth Factor (PDGF) Signaling

Role in Neural Crest Migration: PDGF signaling has been shown to regulate the migration and proliferation of cranial neural crest cells.

Hedgehog Signaling

Role in Neural Crest Migration: While the Hedgehog pathway is mostly known for its roles in patterning and differentiation, there's evidence that it plays a role in the migration of certain neural crest populations, especially trunk neural crest cells.

These pathways, often acting in concert, ensure the precise movement of neural crest cells to their final destinations where they differentiate into diverse cell types. The tight regulation and integration of these pathways are essential for the proper development of structures and tissues derived from the neural crest.

Regulatory Codes for Neural Crest Migration

Neural crest cells (NCCs) are a transient and highly migratory cell population that gives rise to a wide variety of cell types and structures in vertebrates. The migration and differentiation of neural crest cells are coordinated by an intricate network of regulatory codes, which encompass both genetic and epigenetic mechanisms, as well as signaling pathways. Here are the primary regulatory codes responsible for controlling neural crest migration:

Transcriptional Regulation

Neural Crest Specifiers: A cohort of transcription factors, including Snail, Slug (also known as Snail2), FoxD3, Sox9, and Sox10, play pivotal roles in specifying the neural crest lineage and promoting epithelial-to-mesenchymal transition (EMT) which is essential for their migration.

Epigenetic Regulation

Histone Modifications: As mentioned previously, modifications like histone acetylation and methylation can activate or repress gene expression, influencing neural crest formation and migration.
DNA Methylation: Dynamic changes in DNA methylation patterns are crucial for the induction and migration of neural crest cells.
Non-coding RNAs: MicroRNAs (miRNAs) and Long Non-Coding RNAs (lncRNAs) modulate the expression of key genes involved in neural crest development and migration.

Post-translational Modifications

Ubiquitination and SUMOylation: These are processes by which proteins are tagged for degradation or activity alteration. They play a role in the modulation of protein levels and activities related to neural crest migration.

Signaling Pathways

Various pathways, such as BMP, Wnt, FGF, Ephrin-Eph, and Notch, among others, as discussed in the previous section, are crucial for guiding neural crest cells during their migratory journey.

Cell-Cell and Cell-Matrix Interactions

Cadherins: These are cell adhesion molecules. N-cadherin and cadherin-11, for instance, play roles in neural crest cell migration by modulating cell-cell adhesion properties.
Integrins: These are receptors that mediate cell-extracellular matrix interactions. They facilitate neural crest cell migration by binding to specific extracellular matrix components.

Feedback and Feedforward Loops

Regulatory Networks: Interactions between different transcription factors and signaling molecules often result in feedback or feedforward loops, ensuring the tight regulation of neural crest cell migration and differentiation.

External Environmental Cues

Chemotaxis: Gradients of signaling molecules guide neural crest cells to their destinations, with cells moving toward areas of higher ligand concentration.
Contact Inhibition of Locomotion: When neural crest cells collide with one another during migration, they tend to change direction. This phenomenon helps in dispersing the migrating cells.

Cell Polarity and Cytoskeletal Dynamics

Rho GTPases: Proteins like RhoA, Rac1, and Cdc42 regulate the actin cytoskeleton, ensuring proper cell shape and motility during neural crest migration.

The journey of neural crest cells from their origin in the dorsal neural tube to their diverse destinations throughout the embryo is governed by a multifaceted array of regulatory codes. These codes ensure the proper spatiotemporal migration and differentiation of neural crest cells, enabling the formation of many essential structures in vertebrates.

Evidence Supporting Evolutionary Emergence of Neural Crest Migration

The evolutionary emergence of neural crest migration is a topic of great intrigue, particularly when considering the intricate interplay of codes, languages, signaling pathways, and proteins that govern this process. The sophisticated coordination required for neural crest cells to migrate and differentiate poses significant challenges to a purely gradualist model of evolutionary development. The complexity of neural crest migration is evident in the multifaceted regulatory mechanisms involved. For one, the process requires a precise temporal and spatial activation of specific genes. This is controlled by transcription factors, epigenetic markers, and post-translational modifications, all of which need to function in harmony. Any slight deviation in this coordinated dance could lead to developmental anomalies. How could such intricate systems evolve step by step when even a minor disruption can render the whole system non-functional? Similarly, the signaling pathways guiding neural crest migration are highly integrated. Take, for instance, the BMP, Wnt, FGF, and Notch pathways. Each of these interacts with the others, often in complex feedback loops. A change in one pathway can reverberate through the entire system. It is difficult to envision how such interdependent systems could have evolved independently and then somehow merged into a functional whole. Furthermore, the neural crest cells themselves are a marvel of cellular engineering. These cells undergo an epithelial-to-mesenchymal transition, a dramatic change in cellular behavior, allowing them to migrate. This transition involves changes in cell adhesion molecules, activation of specific signaling pathways, and a reorganization of the cytoskeleton. Again, the precise orchestration of these events is crucial; any misstep can result in cells that are either stuck in place or move uncontrollably. Moreover, the codes and languages that guide these processes are analogous to highly sophisticated software programs. Just as a computer program requires a programmer, one might argue that these biological "programs" suggest the work of an intelligent designer. The precision, specificity, and complexity of these codes, which are essential for neural crest migration, seem to defy a piecemeal approach to their development. It could also be highlighted that intermediate stages of such a system might be non-functional. A half-formed signaling pathway or a partially developed transcriptional code would not serve a functional purpose. Without a clear benefit to the organism, these non-functional or sub-functional stages would not be favored by natural selection. The proteins involved in neural crest migration are the workhorses that execute the instructions laid out by the genetic and epigenetic codes. The formation, folding, and function of these proteins are incredibly precise. Even minor changes in a protein's structure can render it non-functional. The simultaneous emergence and coordination of multiple proteins required for neural crest migration is a statistical and evolutionary enigma. The incredible intricacy and interdependence of the systems governing neural crest migration challenge a gradual, stepwise model of evolutionary development. The precision, coordination, and specificity required for this process to function correctly seem to suggest that it arose as a fully-formed, operational system. This perspective aligns with the notion that such a marvel of biological engineering could be the work of an intelligent designer.

Irreducibility and Interdependence of Neural Crest Migration

The intricacy of neural crest migration stands as a testament to the complexity and sophistication of cellular processes. Within this migration, various systems—be it manufacturing, signaling, or regulatory—are deeply interwoven, presenting an image of irreducibility and interdependence that poses challenges to conventional evolutionary narratives.

Manufacturing System: The manufacturing of the cellular machinery and structures that facilitate neural crest cell (NCC) migration is essential. This includes the production of adhesion molecules, receptors, and enzymes. A disruption in the manufacturing of just one of these components can halt the entire migration process, emphasizing the system's irreducible nature.
Signaling Pathways: These pathways, like Wnt, FGF, and Notch, to name a few, guide neural crest migration. They operate in an integrated manner, often relying on feedback and crosstalk to adjust and refine their actions. A partially formed signaling pathway or one missing a critical component would be akin to a broken compass, leading the cell astray.
Regulatory Codes: Transcription factors, epigenetic markers, and other regulatory molecules determine when and where genes are activated or silenced. These codes control everything from the initial specification of the neural crest lineage to the final differentiation of the migrated cells. Without the precise coordination offered by these regulatory codes, neural crest cells would be directionless, proliferating, migrating, or differentiating at the wrong times or places.

Now, how do these systems interact?

Communication and Crosstalk: The various signaling pathways do not operate in isolation. For example, the Wnt pathway often interacts with the Notch pathway, with one modulating the other's activity. This crosstalk ensures that the cell receives consistent instructions from its environment. Furthermore, the manufacturing system produces the molecules, like receptors or enzymes, that the signaling pathways rely on, illustrating the tight interdependence between manufacturing and signaling.
Interdependence with Regulatory Codes: Regulatory codes often control the activity of signaling pathways. A transcription factor might activate a particular receptor or enzyme essential for a signaling pathway, making the signaling and regulatory systems deeply interdependent.

Such a complex dance of interdependence and crosstalk makes it challenging to envision a piecemeal evolutionary development. A half-formed signaling pathway, a partial regulatory code, or a manufacturing system missing a key component would likely result in a non-functional or even detrimental outcome. Without a clear benefit or functionality at every intermediate stage, it's hard to see how these systems could have been favored by natural selection.
Furthermore, the languages these systems employ, whether they're genetic codes, signaling cascades, or epigenetic markers, need to be perfectly in sync for the cell to function correctly. The precision and coordination required for these languages to communicate effectively with one another resemble sophisticated programming, which, to many, suggests the handiwork of an intelligent designer.

In conclusion, the irreducibility and interdependence seen in the systems governing neural crest migration, their communication methods, and the precision with which they operate paint a picture of a masterfully crafted cellular process. The challenges posed by trying to fit this intricate dance into a stepwise evolutionary model further underscore the marvel of neural crest migration.

Neural Crest Migration's Interactions with Other Systems

Neural crest migration is a complex and dynamic process that doesn't function in isolation. Instead, it interacts with a variety of other intra and extracellular systems, revealing a deeply interconnected web of biological processes. Here's a glimpse into some of the systems with which neural crest migration interacts:

Extracellular Matrix (ECM): The ECM is not just a passive support structure. It provides essential guidance cues for migrating neural crest cells (NCCs). Interactions with ECM components like fibronectin, laminins, and collagens can dictate the direction and speed of NCC movement. Furthermore, enzymes that degrade the ECM, such as matrix metalloproteinases, facilitate the migration by clearing pathways.
Growth Factors and Cytokines: These molecules, present in the cellular environment, can either promote or inhibit NCC migration. For instance, growth factors like FGFs and TGF-betas can influence the migration pathways of NCCs.
Cell-Cell Adhesion Systems: NCCs need to detach from their neighboring cells to migrate, a process regulated by molecules like cadherins. However, during migration, transient cell-cell interactions with other migrating NCCs or with stationary cells in their path can also guide their journey.
Paracrine Signaling: Cells in the vicinity of NCCs can secrete signaling molecules that either attract or repel the migrating cells. This paracrine signaling helps to ensure that NCCs reach their intended destinations.
Vascular System: Blood vessels can serve as highways for migrating NCCs, providing both a physical substrate for migration and secreting molecules that guide NCCs.
Nervous System: As NCCs give rise to various components of the peripheral nervous system, their migration is often closely associated with the growth and guidance of axons. Axonal pathways can provide tracks for NCCs to follow.
Endocrine System: Hormones can impact NCC migration. For example, glucocorticoids can influence the differentiation and migration of some NCC populations.
Immune System: There's emerging evidence that immune cells and molecules can influence NCC migration. For example, certain chemokines, which are traditionally viewed as immune signaling molecules, can affect the direction and efficiency of NCC migration.

The aforementioned interactions underscore the fact that neural crest migration is not a standalone event. Instead, it's a process deeply embedded within a network of cellular systems, each influencing and being influenced by the migrating NCCs. This intricate dance of interactions ensures that NCCs reach their destinations, differentiate appropriately, and contribute to the formation of diverse structures in the developing organism. The interconnectedness of these systems emphasizes the complexity and precision required for proper embryonic development.

Premise 1: Systems that rely on intricate semiotic code, languages, and interdependent interactions to function optimally often indicate design in known experiences (e.g., software systems, human-made machinery).
Premise 2: Neural crest migration and its interactions with various cellular systems display an intricate use of semiotic code, languages, and are deeply interdependent, requiring simultaneous and precise orchestration for proper embryonic development.
Conclusion: Given the complexity, precision, and interlocked nature of the systems involved in neural crest migration, it points to a designed setup, akin to our known experiences with intentionally designed complex systems.



Last edited by Otangelo on Fri Sep 01, 2023 4:02 am; edited 1 time in total

https://reasonandscience.catsboard.com

6Evolution of the brain Empty Re: Evolution of the brain Fri Sep 01, 2023 4:00 am

Otangelo


Admin

Neural plate folding and convergence

The development of the central nervous system (CNS) in vertebrates is a complex and meticulously orchestrated process. One of the initial and pivotal stages in the formation of the CNS is the emergence of the neural plate.

What is the Neural Plate?

The neural plate is a thickened layer of ectodermal tissue located in the dorsal region of the early embryo. It represents the primitive precursor to the entire central nervous system, which includes both the spinal cord and the brain.

Function and Development of the Neural Plate:

Induction: The formation of the neural plate, a process termed neural induction, is influenced by signals from the underlying mesoderm, particularly from an area called the notochord. This signaling suppresses the ectoderm's default pathway of becoming epidermis and instead induces it to form the neural plate.
Morphogenesis: Following its induction, the neural plate undergoes significant morphological changes. Its central region starts to elevate, forming the neural folds, while the middle part of the plate sinks, giving rise to the neural groove.
Neurulation: The neural plate's lateral edges (the neural folds) continue to elevate and eventually converge and fuse, transforming the plate into a tubular structure called the neural tube. This process is known as neurulation. The neural tube subsequently gives rise to the spinal cord and brain.

The neural plate's formation and subsequent folding into the neural tube are crucial for proper CNS development. Any perturbations during these processes can result in severe neural tube defects, such as spina bifida or anencephaly.


Neural plate folding and convergence

During early embryonic development, the nervous system begins its formation as a simple, flat structure called the neural plate. As development progresses, this plate undergoes a series of coordinated folding and convergent movements to form the neural tube, a precursor to the spinal cord and brain.

Induction of the Neural Plate: Early in development, signaling molecules induce a portion of the ectoderm (outermost germ layer) to differentiate into the neural plate. This region thickens and elongates.
Neural Fold Formation: As the neural plate continues to elongate, its lateral edges begin to elevate, forming the neural folds.
Convergence and Fusion: The neural folds approach each other at the midline and eventually fuse, transforming the once flat neural plate into a cylindrical neural tube. This tube will eventually give rise to the central nervous system: the spinal cord and brain.
Closure of the Neural Tube: The tube typically closes in multiple regions simultaneously. Any failure in this closure process can lead to neural tube defects, such as spina bifida or anencephaly, depending on where the closure fails.

Importance in Biological Systems

The neural plate folding and convergence process is pivotal for proper nervous system development. The formation of the neural tube is the embryonic foundation for the entire central nervous system. Mistakes or disruptions during this process can lead to severe congenital conditions that can affect an individual's quality of life or even be life-threatening. This complex morphogenetic event showcases the precision required in developmental processes and how tightly regulated and choreographed cellular behaviors are essential for forming complex structures in higher organisms.

How does the neural plate accurately fold and converge to form the neural tube?

The formation of the neural tube from the neural plate is a fundamental process during the embryonic development of many animals, including humans. This process is termed "neurulation." Here is a simplified overview of how the neural plate folds to form the neural tube:

Establishment of the Neural Plate: Early in embryonic development, specific signaling molecules, like bone morphogenetic proteins (BMPs) and their antagonists, help establish the neural plate, a thickened area of the ectoderm (the outermost germ layer).
Neural Plate Border Formation: The edges of the neural plate, known as the neural plate border, become identifiable. The cells here will give rise to the neural crest cells.
Elevation of the Neural Folds: As development progresses, the lateral edges of the neural plate start to elevate and form the "neural folds."
Convergent Extension: Cells in the neural plate undergo changes in their shape and arrangement, a process known as convergent extension. This causes the neural plate to narrow and elongate, pushing the neural folds upwards and towards the midline.
Medial Hinge Point (MHP) Formation: Cells in the center of the neural plate, particularly at the future site of the dorsal midline, become anchored and form a hinge known as the medial hinge point (MHP). This hinge is critical for the bending of the neural plate.
Bending of the Neural Plate: Cells at the MHP change shape, becoming wedge-shaped. This shape change, along with convergent extension, allows the neural folds to move toward each other.
Closure of the Neural Tube: Eventually, the elevated neural folds meet and fuse at the midline, transforming the neural plate into a closed neural tube. The neural tube will give rise to the central nervous system (brain and spinal cord).
Neural Crest Cell Migration: After the neural tube closes, neural crest cells, which are located at the junction between the neural tube and the non-neural ectoderm, begin to migrate and differentiate into various cell types, such as peripheral neurons, glial cells, and melanocytes.
Closure Completion: The process of neural tube closure starts at multiple points along the anterior-posterior axis and proceeds bidirectionally. For instance, in humans, the neural tube closure starts in the region of the future neck and proceeds both cranially (toward the head) and caudally (toward the tail).

How does this convergence contribute to the formation of the CNS?

The convergence and folding of the neural plate to form the neural tube is the foundational process in the formation of the central nervous system (CNS), which consists of the brain and the spinal cord. Here's how this convergence contributes to the formation of the CNS:

Specification of CNS Regions: As the neural plate folds and converges to form the neural tube, various regions of the tube become specified to give rise to different parts of the CNS. The anterior (front) part of the neural tube will develop into the brain, while the posterior (back) part will become the spinal cord.
Neural Tube as the Precursor: The neural tube itself serves as the precursor to the CNS. Once it has formed, its internal cavity will become the ventricular system of the brain and the central canal of the spinal cord.
Patterning and Differentiation: Within the neural tube, a variety of molecular gradients and signaling pathways define specific regions along both the anterior-posterior axis (from head to tail) and the dorsal-ventral axis (from back to belly). These signaling pathways allow for the differentiation of specific types of neurons and glial cells in precise locations. For instance, motor neurons develop in the ventral part of the neural tube, while sensory neurons develop from the neural crest cells that originate from the dorsal part of the tube.
Brain Vesicle Formation: In the anterior part of the neural tube, it will start to expand and differentiate further, giving rise to three primary vesicles: the forebrain (prosencephalon), midbrain (mesencephalon), and hindbrain (rhombencephalon). As development continues, these vesicles will undergo further specialization to form the different regions of the brain.
Growth and Elaboration: As the neural tube matures, there's a rapid proliferation of neurons. Neurons then extend axons and dendrites, forming intricate networks of connections. Glial cells also proliferate and play roles in supporting neurons, insulating axons, and maintaining homeostasis.
Central Canal Formation: The lumen (internal cavity) of the neural tube persists as the central canal in the spinal cord and as the ventricular system in the brain. These cavities will be filled with cerebrospinal fluid, which plays a crucial role in cushioning the CNS, providing nutrients, and removing waste.

Evolution of the brain Sem_t109
Neural crest formation during neurulation. 1

At what point in the evolutionary timeline did neural plate folding and convergence appear?

The process of neurulation, which involves the folding and convergence of the neural plate to form a neural tube, is observed in chordates, a large and diverse group of animals that includes vertebrates (animals with backbones, like fish, birds, mammals, etc.) as well as some invertebrates, such as tunicates and cephalochordates (e.g., amphioxus or lancelets). Given this distribution, the appearance of neurulation is claimed to be traced back to the common ancestor of chordates. This would suggest that the process is quite ancient, originating more than 500 million years ago during the Cambrian period or even earlier. The Cambrian period, in particular, is notable for the "Cambrian explosion," a relatively short evolutionary interval during which many major animal phyla (including chordates) appeared in the fossil record.

Simpler Nervous Systems Before Neurulation: Before the evolution of chordates, simpler nervous systems would have existed in other animal groups. For instance, cnidarians (like jellyfish) and flatworms have nerve nets or simple nerve cords, but they do not form via neurulation.
Diversification of Neural Structures: Within chordates, the specific structure and complexity of the central nervous system (CNS) would have diversified. While all chordates form a neural tube, the subsequent development of this tube can vary widely. For example, vertebrates possess a much more complex CNS compared to tunicates or lancelets.
Vertebrate Advancements: Among the chordates, vertebrates represent a further elaboration on the neural tube theme. The vertebrate CNS (brain and spinal cord) is considerably diversified and specialized, leading to the advanced brains observed in mammals, birds, reptiles, and other groups.

What genetic information was necessary to be created de novo, to instantiate neural plate folding and convergence?

Neural plate folding and its subsequent convergence to form the neural tube are fundamental processes during vertebrate embryogenesis. This process leads to the formation of the central nervous system, including the brain and the spinal cord. The precise molecular and genetic pathways that guide this process have been an area of intense research, with many genes and signaling pathways implicated.
It is essential to differentiate between genes that were already present in an ancestor and used in a new context versus those that might have arisen de novo (from scratch).  Here's a basic outline of some of the main genetic players involved in neural plate folding and convergence, although it's worth noting that this is an extensive topic and the list isn't exhaustive:

Induction of the Neural Plate: Before folding, there's neural induction, which delineates the neural plate from the surrounding ectoderm. The process is thought to be driven by the suppression of BMP signaling by factors like Noggin, Chordin, and Follistatin that are secreted from the underlying organizer tissue.
Boundary Formation: The edges of the neural plate, called the neural plate border, become defined. PAX3, PAX7, MSX1/2, and ZIC1 are some of the genes that define this region and subsequently give rise to neural crest cells.
Convergence and Extension Movements: Cellular movements cause the neural plate to narrow (convergence) and lengthen (extension). This involves non-canonical Wnt signaling with players like Wnt11 and the PCP (planar cell polarity) pathway. Key components include Vangl2, Prickle, and Celsr1.
Neural Plate Folding: Cells in the medial hinge point (MHP) undergo apical constriction driven by actin-myosin contractility, causing the neural plate to fold. Shh (Sonic Hedgehog) signaling is essential for MHP specification.
Closure of the Neural Tube: The elevated edges of the neural plate, called the neural folds, converge and fuse at the dorsal midline. E-cadherin, a cell-cell adhesion molecule, is crucial for this fusion.
Other Signaling Pathways: A multitude of other signaling pathways, including retinoic acid, FGF, and Notch, have roles in various aspects of neural plate development and neural tube closure.

The genes and pathways mentioned above did not all arise de novo for the purpose of neural tube formation. Many are claimed to have been co-opted from other processes and have roles in other parts of embryonic development.  The genetics of neural tube development is complex, and much has been learned from model organisms such as the frog, zebrafish, chickens, and mouse. However, many details, especially at the molecular and cellular level, are still the subject of active research.

Which manufacturing codes and languages had to emerge and be employed for neural plate folding and convergence?

When we talk about "manufacturing codes" and "languages" in the context of biology and embryogenesis, we're using metaphorical language to describe the complex interplay of genetic, molecular, and cellular processes that drive development. There are specific sequences, signals, and regulatory mechanisms that guide development. These biological "codes" are carried out through gene expression, protein-protein interactions, and cell-cell communication.

Genetic Code: This is the actual sequence of DNA that encodes genes. Every cell in an organism (with some exceptions like mature red blood cells) contains the entire genetic code of that organism. Genes get transcribed into RNA and then many of them get translated into proteins, which carry out most of the functions in cells.
Regulatory Elements: Beyond the genes themselves, the DNA contains regulatory sequences that determine when, where, and how strongly a gene gets expressed. These include promoters, enhancers, silencers, and insulators. Transcription factors bind to these regions to activate or repress gene expression. This can be thought of as the "programming logic" of development.
Signaling Pathways: Cells communicate with each other using signaling molecules. A cell will produce and release a signaling molecule (e.g., a growth factor) which will bind to a receptor on another cell, initiating a cascade of events inside that cell that can change its behavior. This is akin to a "communication protocol" between cells.
Feedback Loops: Many biological processes involve feedback mechanisms where the product of a process affects the rate of that process. This can result in systems that are self-regulating, oscillating, or that have multiple stable states. This is a kind of "dynamic programming."
Cell Behaviors: Cells can move, change shape, divide, differentiate, or die. These behaviors are the result of interpreting the genetic "code" in a specific context. For example, neural plate cells undergo specific movements and changes in shape that lead to folding and convergence.

In the specific context of neural plate folding and convergence, all of these "codes" and "languages" come into play. The genetic code provides the raw information, regulatory elements determine the timing and location of gene expression, signaling pathways allow cells to coordinate their behavior, molecular machines carry out the functions, feedback loops ensure robustness and precision, and the end result is a set of coordinated cell behaviors that form the neural tube.

What epigenetic regulatory mechanisms are necessary for proper neural plate folding and convergence?

DNA Methylation: This is the addition of a methyl group to the cytosine base in DNA. In general, methylation of gene promoter regions is associated with repression of gene expression. DNA methylation patterns change dynamically during neural development and play a role in cellular differentiation and maintaining the identity of neural cells.
Histone Modifications: Histones are proteins around which DNA is wrapped, forming a structure called nucleosome. The tails of histones can be chemically modified in various ways, such as methylation, acetylation, phosphorylation, and ubiquitination. Each modification can have different effects on gene expression, depending on the specific histone, the amino acid modified, and the type of modification. For example, histone H3 lysine 27 trimethylation (H3K27me3) is associated with gene repression, while histone H3 lysine 4 trimethylation (H3K4me3) is linked with gene activation. These modifications play a role in defining and maintaining cellular identities during neural development.
Chromatin Remodeling: Chromatin is the complex of DNA and histones. Chromatin remodeling complexes can change the positioning or composition of nucleosomes, affecting the accessibility of DNA to transcriptional machinery. This can either activate or repress gene expression, depending on the context. For instance, the SWI/SNF (or BAF) complex has been implicated in neural development and differentiation.
Non-coding RNAs: These are RNA molecules that don't code for proteins but play roles in regulating gene expression. Two significant classes are microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). miRNAs can bind to messenger RNAs (mRNAs) and prevent their translation or lead to their degradation. lncRNAs have diverse functions, including serving as scaffolds for protein complexes, sequestering miRNAs, and directly interacting with DNA to affect its structure and accessibility. Several non-coding RNAs are crucial for various aspects of neural development.
RNA Methylation: Just as DNA can be methylated, modifications to RNA, such as N6-methyladenosine (m6A), have been discovered and play roles in RNA stability, splicing, and translation. These modifications can impact neural development, although the full scope of their functions is still being explored.
Three-dimensional Chromatin Organization: The spatial arrangement of chromatin in the nucleus, forming loops and domains, plays a role in gene regulation by bringing distant regulatory elements into proximity with genes. Structures like topologically associating domains (TADs) and the associated proteins (like CTCF) that help shape the 3D genome can influence neural gene expression patterns.

These epigenetic mechanisms interact and often converge at particular genes to ensure precise spatial and temporal gene expression patterns necessary for neural plate folding, convergence, and other aspects of neural development. Dysregulation of these mechanisms can lead to developmental disorders or diseases later in life.

Which signaling pathways support neural plate folding and convergence?

The formation of the neural tube from the neural plate involves intricate cellular processes that are tightly regulated by various signaling pathways. These pathways guide the neural plate cells in their migration, proliferation, differentiation, and morphological changes. Here are the major signaling pathways implicated in neural plate folding and convergence:

Bone Morphogenetic Protein (BMP) Pathway: During neural induction, the inhibition of BMP signaling is critical for neural plate formation. Molecules like Noggin, Chordin, and Follistatin, which are secreted from the underlying organizer tissue (such as the notochord), act as BMP antagonists. In the absence of BMP signaling, ectodermal cells are more inclined to adopt a neural fate.
Sonic Hedgehog (Shh) Pathway: Shh is secreted primarily from the notochord and later from the floor plate of the developing neural tube. It plays a role in specifying ventral cell fates within the neural tube and is essential for the formation and function of the medial hinge point during neural plate folding.
Wnt Pathway: The Wnt signaling pathway is implicated in various aspects of neural development. Canonical Wnt/β-catenin signaling plays a role in dorsal neural tube cell fates, whereas non-canonical Wnt signaling (often referred to as the Planar Cell Polarity or PCP pathway) is involved in the convergence and extension movements that narrow and elongate the neural plate.
Notch Pathway: Notch signaling is pivotal for cell-cell communication and plays roles in regulating neural progenitor differentiation and maintaining the balance between neural progenitors and differentiated neurons.
Fibroblast Growth Factor (FGF) Pathway: FGF signaling is involved in neural induction and the subsequent patterning of the neural plate. It also plays roles in neural progenitor proliferation and differentiation.
Retinoic Acid (RA) Pathway: RA, a derivative of vitamin A, plays roles in the anteroposterior patterning of the neural tube, especially in the hindbrain region. It also affects the timing of neurogenesis.
TGF-β/Activin/Nodal Pathway: Members of the TGF-β superfamily, like Activin and Nodal, are involved in mesendoderm formation and can influence neural induction and patterning indirectly through their effects on organizer tissue formation.
Cilia-Associated Signaling: Primary cilia, which are small microtubule-based protrusions from the cell surface, play roles in sensing and transducing signals from various pathways, including Shh and Wnt. Proper ciliary function is required for correct Shh signal transduction during neural development.

Many of these pathways interact, and cells often integrate signals from multiple pathways to make decisions about fate and behavior. Proper coordination and regulation of these pathways are essential for the correct morphogenesis of the neural tube, and disruptions can lead to neural tube defects, a common class of congenital malformations.

What regulatory codes are essential for the maintenance and operation of neural plate folding and convergence?

The process of neural plate folding and convergence is a complex orchestration of cellular behaviors governed by intricate regulatory networks. These networks can be thought of as "regulatory codes" that ensure cells exhibit the right behavior at the right time and place. Here are the primary regulatory codes essential for neural plate folding and convergence:

Transcriptional Regulation: This refers to the control of gene expression at the level of transcription. Specific sets of transcription factors are expressed in the neural plate and its border, and they regulate the genes responsible for neural identity, morphogenesis, and other processes associated with neural plate development.
Key factors: Sox2, Sox3, Zic1/2/3, Pax3/4/6, and Msx1/2 are some of the transcription factors crucial for neural plate and neural crest specification, respectively.
Post-Transcriptional Regulation: After genes are transcribed, their RNA products can still be regulated, affecting their stability, splicing, or translation.
Role of microRNAs (miRNAs): These are small non-coding RNAs that can bind to messenger RNAs (mRNAs) and inhibit their translation or induce their degradation. Specific miRNAs have roles in various aspects of neural development.
Epigenetic Regulation: As discussed previously, modifications like DNA methylation, histone modifications, and chromatin remodeling play a role in determining which genes are accessible and can be transcribed. For example, the repressive H3K27me3 mark may be placed on non-neural genes in the neural plate to ensure they remain off.
Cell-Cell Signaling Pathways: Cells in the developing embryo communicate with each other, sending and receiving signals that dictate cell behaviors. As mentioned before, BMP, Shh, Wnt, FGF, Notch, and other pathways are all active during neural plate development, providing necessary regulatory inputs.
Mechanical Forces and Feedback: The physical properties of cells and their environment can influence and be influenced by the genetic and signaling codes. For instance, the apical constriction of cells in the neural plate, driven by actin-myosin contractility, is essential for neural tube folding. Feedback from these mechanical processes can further influence gene expression and cellular behaviors.
Cell Adhesion and Polarization: The proper adhesion between cells and the polarization of cells (distinguishing an apical from a basal side) are crucial for the coordinated movements during neural plate folding. Molecules like cadherins and integrins play a role in ensuring cells stick together appropriately, and proteins like Par3, Par6, and aPKC help establish and maintain cell polarity.
Feedback Loops: Several signaling pathways have built-in feedback mechanisms, both positive and negative. For example, a signaling molecule might activate the transcription of its inhibitor, creating a negative feedback loop. These loops ensure robustness, fine-tuning, and can create dynamic behaviors like oscillations.
Gradient and Threshold Interpretation: Many signaling pathways operate as gradients across tissues. Cells can interpret the level (or concentration) of a signal and respond accordingly, often by expressing different genes above specific thresholds. This mechanism helps establish different cell fates across a tissue.

The integration of these various codes, from the genetic level to the physical interactions between cells, ensures the accurate morphogenesis of the neural tube. Dysregulation of any part of this regulatory network can lead to neural tube defects and other developmental anomalies.

Is there scientific evidence supporting the idea that neural plate folding and convergence were brought about by evolution?

Neural plate folding and convergence is a marvel of biological engineering, showcasing an intricate and interwoven dance of cellular behaviors, signaling cascades, genetic codes, and molecular mechanisms. When delving into the specifics of this process, one might argue that the complex series of events leading to the formation of the neural tube appears orchestrated in such a way that partial or intermediate systems  seem non-functional or even detrimental. 

Complex Interdependence: The process of neural plate folding and convergence isn't a simple one-step mechanism. It requires the intricate coordination of multiple systems, each consisting of numerous components. For instance, signaling pathways like BMP, Shh, and Wnt must work in tandem, where the absence or malfunction of one pathway could disrupt the entire developmental process. This raises the question: how could such a multifaceted system have evolved piece by piece if each component is reliant on the others for function?
Requirement of Precise Timing: The sequence and timing of events during neural plate development are crucial. A delay in one process or an early initiation of another could lead to catastrophic consequences, such as neural tube defects. For evolution to guide such a finely-tuned process step by step seems a challenging proposition given the precision required.
No Advantage in Partial Systems: For evolution to favor a trait, that trait usually needs to confer some advantage. However, with neural plate folding, partial or intermediate stages might not provide any functional benefit. For instance, a partially folded neural plate that doesn't close might not be beneficial and could instead be harmful.
Instantiation of Genetic and Epigenetic Codes: The genetic code guiding neural plate development is intricate, encompassing not just genes but also regulatory elements, enhancers, silencers, and more. Beyond genetics, there's also the layer of epigenetics, which determines which genes are accessible and when. Such multilayered regulation seems too complex to have arisen step by step without a guiding force, as missing a single component could render the entire system non-functional.
Proteins and Their Specificity: Many proteins involved in neural plate folding are multifunctional, playing roles in various cellular processes. Their precise interactions, binding sites, and functions in the context of neural plate development are specific and intricate. The chance of emergence of such multifunctional proteins seems unlikely, especially since a minor alteration in their structure could render them non-functional or even harmful.
Cellular Behaviors and Coordination: Beyond molecular and genetic intricacies, there's the added complexity of cellular behaviors like migration, proliferation, and differentiation. Each cell in the developing neural plate seems to know its role, suggesting a pre-defined plan rather than a series of evolutionary accidents.

Potential for Detriment in Errors: In many biological systems, small errors can be tolerated. However, in processes as complex and vital as neural tube formation, mistakes can lead to severe consequences, like spina bifida or anencephaly. Given the high stakes, the evolution of such a system in a piecemeal manner seems improbable, as errors would be catastrophic.

In light of the above points, one might argue that the complexity and precision required for neural plate folding and convergence make a step-by-step evolutionary process implausible. Instead, it might be posited that the entire system, in all its intricacy, had to emerge fully formed and functional, suggesting a design by an intelligent entity.

What components of neural plate folding and convergence are irreducible and interdependent for its proper function?

For neural plate folding and convergence, several components and processes are critical, and they function in an interdependent manner. 

Signaling Pathways: A variety of signaling pathways, including BMP, Shh, Wnt, and FGF, must be tightly coordinated for neural plate folding. The absence or malfunction of one pathway can disrupt the balance of signals required for proper neural plate development.
Transcription Factors: These are proteins that help turn specific genes on or off. Many transcription factors are crucial for neural plate development, including Sox2, Sox3, Pax3, and Zic genes. Their absence or malfunction can impede proper neural differentiation and folding.
Mechanical Forces: Cellular behaviors, such as apical constriction, are driven by actin-myosin contractility. This mechanical force is critical for the bending of the neural plate. Without it, neural plate folding would not occur.
Cell-Cell Adhesion Molecules: Molecules like cadherins are crucial for cells to adhere to each other during the folding process. Disruption of these adhesion molecules can prevent the neural plate from folding properly.
Cell Polarity Proteins: These ensure that cells have a distinct "top" (apical) and "bottom" (basal) side. Proteins like Par3, Par6, and aPKC are crucial for establishing this polarity. Without proper cell polarity, the coordinated behaviors required for neural plate folding would be disrupted.
Extracellular Matrix (ECM): The ECM provides structural support to tissues. Changes in ECM composition or the interaction of cells with the ECM can influence neural plate folding. Molecules like fibronectin play a role in guiding cell movements during this process.
Apoptosis Mechanisms: Programmed cell death, or apoptosis, can shape the neural plate and tube. Properly timed apoptosis is essential for neural tube closure in some regions.
Feedback Loops: Signaling pathways often have feedback mechanisms to ensure they aren't perpetually on or off. These feedback loops, both positive and negative, are crucial for the dynamic behaviors observed during neural plate folding.

In a system as intricate as neural plate folding and convergence, each component and process has a role to play, and they all work in concert. From an "irreducible complexity" perspective, one could argue that removing any of these components would hinder or halt the process altogether. However, it's worth noting that the concept of irreducible complexity is controversial in the broader scientific community, as many believe that evolutionary processes can, and often do, build complex systems incrementally over time.

Once neural plate folding and convergence is operational, with what intra and extracellular systems does it interact?

The process of neural plate folding and convergence does not occur in isolation. Instead, it interacts with various intracellular and extracellular systems that coordinate to ensure the proper formation of the neural tube. Here's a breakdown of these interactions:

Intracellular Systems: Cytoskeletal Dynamics: The cytoskeleton, composed primarily of actin filaments, microtubules, and intermediate filaments, undergoes dynamic rearrangements during neural plate folding. Particularly, actin-myosin contractility at the apical side of the neural plate cells drives their shape changes, which are critical for the folding process.
Cell Polarity Machinery: Intracellular polarity complexes, such as the Par complex (including Par3, Par6, and aPKC), help establish and maintain the apical-basal polarity of the neural plate cells. Proper cell polarity is essential for the coordinated cell behaviors during neural plate folding.
Transcriptional and Translational Machinery: The cellular machinery responsible for gene expression is continuously active, ensuring that the right proteins are synthesized at the right time. Transcription factors, ribosomes, and various associated molecules play pivotal roles.
Intracellular Signaling Pathways: These pathways interpret extracellular signals and ensure appropriate cellular responses. Examples include the cascades triggered by BMP, Shh, and Wnt signals, which influence gene expression and cell behavior.

Extracellular Systems

Extracellular Matrix (ECM): The ECM provides structural support and guidance cues for migrating cells during neural tube closure. It contains molecules like fibronectin, laminin, and collagen. Neural plate cells interact with the ECM via cell surface receptors, such as integrins.
Cell-Cell Communication: Neighboring cells communicate via various methods:
Gap Junctions: Allow for direct cytoplasmic communication between cells.
Adherens Junctions and Tight Junctions: Provide mechanical attachment between cells and help maintain tissue integrity.
Morphogens and Growth Factors: These are signaling molecules that can influence cell fate and behavior. They often form gradients in the developing embryo, with cells responding differently based on their position within the gradient. Examples affecting neural development include BMPs, Shh, Wnts, and FGFs.
Surrounding Tissues: The behavior of the neural plate is influenced by neighboring tissues. For example:
Surface Ectoderm: Lies adjacent to the neural plate and influences its behavior through secreted signals and physical interactions.
Mesoderm: Especially the notochord, which lies beneath the neural plate, secretes signals like Shh that influence neural plate development.
Endoderm: The most ventral germ layer can also exert influences on neural plate and tube dynamics.
Mechanical Forces: Forces from neighboring tissues can influence neural plate behavior. For instance, the expansion of the adjacent surface ectoderm can exert forces on the neural plate.

In essence, neural plate folding and convergence are processes deeply integrated with a host of intracellular and extracellular systems, showcasing the remarkable coordination and complexity of embryonic development.

1. Wikipedia: Neural plate

https://reasonandscience.catsboard.com

7Evolution of the brain Empty Re: Evolution of the brain Fri Sep 01, 2023 4:00 am

Otangelo


Admin

Neuronal Pruning and Synaptogenesis

How do neuronal pruning and synaptogenesis regulate neural circuits during development and in response to experience?

Neuronal pruning and synaptogenesis are critical processes that regulate neural circuits during development and in response to experience. These processes shape the intricate network of connections within the brain, allowing it to efficiently process information and adapt to changing environments. Here's how neuronal pruning and synaptogenesis contribute to the regulation of neural circuits:

Neuronal Pruning

Overproduction of Neurons and Connections: During early brain development, there is an overproduction of neurons and synapses. This abundance of connections is important to ensure that the brain has the potential to establish a wide range of circuits.
Competition for Resources: Neurons and synapses compete for limited resources, such as nutrients and trophic factors. This competition leads to the selective survival of the fittest neurons and synapses while eliminating weaker ones.
Synaptic Elimination: Neuronal pruning involves the selective elimination of excess synapses. This process is often guided by neural activity; synapses that are less active are more likely to be eliminated. This activity-dependent pruning helps refine and strengthen the most relevant connections.
Role of Apoptosis: In some cases, the elimination of excess neurons occurs through programmed cell death, or apoptosis. This controlled cell death is a natural part of neural development and helps sculpt the brain's architecture.

Synaptogenesis

Formation of New Synapses: Synaptogenesis is the process by which new synapses are formed between neurons. This process begins early in development and continues throughout life, allowing the brain to adapt to new experiences and learn new information.
Activity-Dependent Wiring: Neural activity plays a crucial role in synaptogenesis. Neurons that fire together establish connections, leading to the strengthening of synapses and the creation of functional circuits. This process is a basis for learning and memory.
Structural and Functional Plasticity: Synaptogenesis contributes to the brain's plasticity – its ability to reorganize itself in response to experience. New synapses can form in response to learning, environmental changes, or sensory input.
Critical Periods: During certain developmental stages, such as critical periods, the brain is particularly sensitive to experience, and synaptogenesis is highly active. These periods are essential for the proper wiring of sensory systems and the development of complex skills.

Neuronal pruning and synaptogenesis work in concert to refine neural circuits by eliminating unnecessary connections and strengthening relevant ones. This dynamic interplay between elimination and formation of synapses is crucial for the development, plasticity, and adaptability of the brain's neural circuits in both early development and throughout life.

How do these processes contribute to the overall functionality and plasticity of the nervous system?

Neuronal pruning and synaptogenesis play pivotal roles in shaping the functionality and plasticity of the nervous system. These processes collectively contribute to the refinement, efficiency, and adaptability of neural circuits, allowing the brain to process information, learn, and respond to experiences in a dynamic manner.

Overall Functionality:

Elimination of Redundant Connections: Neuronal pruning ensures that only the most relevant and effective connections are retained in the neural network. By eliminating redundant or weaker connections, the brain optimizes the transmission of signals and reduces noise, leading to more efficient information processing.
Circuit Specialization: Pruning and synaptogenesis help neural circuits become specialized for specific functions. As connections are refined, distinct circuits dedicated to sensory processing, motor control, memory, and other cognitive functions emerge. This specialization enhances the overall functionality of the nervous system.
Network Balance: Neuronal pruning prevents circuits from becoming overly complex and unwieldy. This maintains a balance between different neuronal populations, preventing an excessive number of connections that could impede efficient information flow.

Plasticity and Adaptability:

Experience-Dependent Changes: Synaptogenesis allows the nervous system to adapt to changing environments and experiences. New synapses can form as a response to learning or exposure to novel stimuli, enabling the brain to incorporate new information into existing networks.
Learning and Memory: The ability of synapses to strengthen or weaken in response to activity, known as synaptic plasticity, underlies learning and memory processes. Long-term potentiation (LTP) and long-term depression (LTD) are forms of synaptic plasticity that contribute to the encoding and retention of information.
Sensory Development: During critical periods in development, synaptogenesis is particularly active, allowing sensory systems to wire themselves in response to specific experiences. This is crucial for the proper development of sensory perception.
Recovery from Injury: Neuronal pruning and synaptogenesis also play roles in recovery after neural injury. The brain can rewire itself to some extent by forming new connections around damaged areas, aiding in functional recovery.

Neurodevelopmental Disorders and Plasticity:

Imbalance and Disorders: Disruptions in neuronal pruning and synaptogenesis can lead to neurodevelopmental disorders. For instance, conditions like autism spectrum disorder (ASD) are associated with altered synapse formation and connectivity.
Therapeutic Potential: Understanding these processes is essential for developing therapies for neurodevelopmental disorders. Promoting adaptive synaptogenesis and modifying pruning patterns could potentially help treat certain conditions.

Neuronal pruning and synaptogenesis are crucial mechanisms that not only refine neural circuits for optimal functionality but also provide the nervous system with the capacity to adapt, learn, and respond to experiences throughout life. These processes are foundational to the brain's remarkable ability to process information, form memories, and continuously reshape itself in response to the world around it.

How do neuronal pruning and synaptogenesis contribute to the overall functionality and plasticity of the nervous system?

Neuronal pruning and synaptogenesis are fundamental processes that play crucial roles in shaping the functionality and plasticity of the nervous system. These processes involve the refinement of neural connections, leading to more efficient neural circuits and adaptive responses. Here's how neuronal pruning and synaptogenesis contribute to the overall functionality and plasticity of the nervous system:

Neuronal Pruning

Exuberant Connection Formation: During early development, neurons form an excessive number of connections, resulting in a dense network of synapses. This exuberant connectivity allows the nervous system to establish a wide range of potential pathways and interactions.
Competition and Refinement: Neuronal activity plays a crucial role in determining which synapses are strengthened and which are weakened. Synapses that are frequently activated are reinforced, while those that are less active are eliminated through a process called synaptic pruning. This competitive process refines neural connections, eliminating unnecessary or weak synapses and enhancing the efficiency of information transmission.
Sculpting Circuitry: Neuronal pruning is responsible for sculpting neural circuits into more precise and functional configurations. This fine-tuning of connections enhances the specificity of neural pathways, allowing for more accurate and efficient signal processing.

Synaptogenesis

Formation of New Synapses: Synaptogenesis involves the formation of new synapses between neurons. This process occurs throughout life, not just during development, and it contributes to learning, memory, and adaptive responses to environmental changes.
Experience-Dependent Plasticity: Synaptogenesis is influenced by experiences and environmental factors. Learning new skills or adapting to new situations often involves the creation of new synapses or the strengthening of existing ones. This experience-dependent plasticity allows the nervous system to adapt and learn from its surroundings.
Neuroplasticity and Recovery: Following injuries or changes in sensory input, synaptogenesis can contribute to the brain's ability to rewire itself and recover lost function. Neurons can establish new connections or alter existing ones to compensate for damage or changes in input.

Neuronal pruning and synaptogenesis are essential processes that optimize the structure and function of the nervous system. Neuronal pruning refines neural connections, while synaptogenesis allows for the formation of new synapses, enabling learning, memory, and adaptive responses. These processes together contribute to the remarkable plasticity and adaptability of the nervous system throughout life.

Evolution of the brain Sem_t108
A model view of the synapse 1

At what point in the evolutionary timeline did neuronal pruning and synaptogenesis first appear?

Neuronal pruning and synaptogenesis are complex processes that are intimately linked to the development and functionality of the nervous system. While the exact point in the evolutionary timeline when these processes first appeared is not definitively known, it's supposed that they emerged gradually as nervous systems became more sophisticated.

The evolution of nervous systems would have been a gradual process that spans millions of years, making it challenging to pinpoint precise stages in which specific mechanisms like neuronal pruning and synaptogenesis emerged. 

Early Nervous System Evolution: In the earliest multicellular organisms, nerve cells (neurons) would have started to form basic networks, allowing for simple sensory and motor responses. These early networks would have lacked the complex pruning and refinement mechanisms seen in more advanced nervous systems.
Emergence of Synaptic Connections: As nervous systems would have become more complex, the formation of synaptic connections would have became more important. Synapses, the junctions between neurons, would have allowed for communication and signal transmission between nerve cells. Over time, mechanisms that promoted the strengthening or weakening of synapses would have emerged to enhance the efficiency of signal transmission.
Refinement and Pruning: As nervous systems would have continued to evolve, mechanisms of neuronal pruning probably would have developed as a way to fine-tune neural connections. This would have been driven by the need for more efficient neural circuits, as well as the optimization of limited resources in the developing organisms.
Adaptation and Plasticity: The ability to form new synapses and adapt existing ones, which is a hallmark of synaptogenesis, would have provided significant evolutionary advantages. Organisms with the ability to adjust their neural circuits based on experiences and environmental changes would have been better equipped to survive and thrive in changing conditions.

What de novo genetic information is thought to have been necessary to instantiate neuronal pruning and synaptogenesis?

The mechanisms underlying neuronal pruning and synaptogenesis involve intricate genetic and molecular processes that regulate the formation, refinement, and elimination of neural connections. While it's not necessarily the case that entirely new genetic information was required to instantiate these processes, the proper orchestration of existing genetic information would have been crucial. Here are some key aspects of genetic information and molecular mechanisms thought to be involved:

Gene Expression and Regulation: Existing genes in an organism's genome are responsible for producing the proteins and molecules necessary for neuronal development and plasticity. The activation or repression of specific genes during different developmental stages is critical for initiating and guiding processes like synaptogenesis and neuronal pruning.
Signaling Pathways: Various signaling pathways, involving proteins and molecules such as growth factors, neurotransmitters, and their receptors, play essential roles in regulating neuronal development and connectivity. These pathways transmit information that guides the formation, strengthening, and elimination of synapses.
Synaptic Activity and Plasticity Genes: Certain genes are associated with synaptic plasticity—the ability of synapses to change their strength in response to activity. These genes, such as those involved in the regulation of neurotransmitter receptors and synaptic structure, contribute to the dynamic nature of synaptogenesis and pruning.
Epigenetic Modifications: Epigenetic modifications, which influence gene expression without altering the underlying DNA sequence, also play a role in neuronal development. These modifications can be influenced by experiences and environmental factors, contributing to the adaptive nature of the nervous system.
Cell-Cell Interactions: Cell adhesion molecules and guidance cues are essential for establishing and refining neural connections. These molecules, guided by genetic information, help neurons find their appropriate partners and form synapses in specific patterns.

The genetic information necessary for neuronal pruning and synaptogenesis involves the coordination of existing genes, signaling pathways, and molecular mechanisms. Rather than requiring entirely new genetic elements, these processes rely on the careful regulation and interaction of existing genetic information to sculpt the intricate neural circuits and adaptability observed in the nervous system.

What manufacturing codes and languages would have had to emerge and be employed for the processes of neuronal pruning and synaptogenesis?

The processes of neuronal pruning and synaptogenesis involve intricate cellular and molecular interactions rather than literal manufacturing codes and languages like those used in human-made technologies. Nevertheless, we can draw an analogy to the concept of "codes" and "languages" in biological terms to describe the molecular instructions and interactions that guide these processes. Here are some analogies to help understand the concept:

Molecular Signaling Pathways: In a metaphorical sense, molecular signaling pathways can be seen as analogous to a "language" that cells use to communicate with each other. Various molecules, such as neurotransmitters, growth factors, and receptors, act as "words" in this cellular communication. Cells "read" these signals to initiate processes like neuronal pruning and synaptogenesis.
Genetic Information and Expression: The genetic code present in an organism's DNA can be likened to a "manufacturing code." Genes contain the instructions for producing proteins and molecules needed for neuronal development and plasticity. The process of gene expression, where DNA is transcribed into RNA and then translated into proteins, can be seen as the "manufacturing" process based on these codes.
Epigenetic Marks and Modifications: Epigenetic modifications, which can influence gene expression without changing the underlying DNA sequence, could be considered as a form of regulatory "coding." These modifications act like switches that turn genes on or off, impacting the course of neuronal development and the dynamics of synaptogenesis.
Cell-Cell Communication: Cell-adhesion molecules and guidance cues can be thought of as a type of "communication language" that cells use to establish proper connections. These molecules guide neurons to their appropriate partners during synaptogenesis and contribute to the spatial organization of neural circuits.

While there aren't literal manufacturing codes and languages involved in neuronal pruning and synaptogenesis, the analogy helps us grasp the complexity of molecular interactions and instructions that guide these processes. The language of molecular signaling, genetic information, epigenetic regulation, and cell-cell communication collectively orchestrates the intricate development and refinement of neural connections in the nervous system.

Which epigenetic regulatory mechanisms are critical for directing neuronal pruning and synaptogenesis?

Epigenetic regulatory mechanisms play a vital role in shaping the processes of neuronal pruning and synaptogenesis by modulating gene expression and influencing the formation and elimination of synapses. Here are some of the critical epigenetic mechanisms involved:

DNA Methylation: DNA methylation involves the addition of methyl groups to specific regions of DNA, typically cytosine residues in CpG dinucleotides. In neuronal development, DNA methylation can influence the expression of genes involved in synaptic plasticity, axon guidance, and cell adhesion. Changes in DNA methylation patterns can lead to lasting alterations in synaptic connectivity.
Histone Modifications: Histones are proteins around which DNA is wound, forming chromatin. Modifications to histone proteins, such as acetylation, methylation, phosphorylation, and more, can influence how tightly DNA is packaged and thus affect gene accessibility. Specific histone modifications are associated with active or repressed gene expression, impacting processes like neuronal pruning and synaptogenesis.
Non-coding RNAs: Non-coding RNAs, such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), can regulate gene expression by binding to target messenger RNAs (mRNAs) and influencing their stability or translation. These RNA molecules can have profound effects on neuronal development, including synapse formation and elimination.
Activity-Dependent Epigenetic Changes: Neuronal activity, such as synaptic stimulation, can trigger epigenetic modifications that influence gene expression. For example, neuronal activity can lead to changes in DNA methylation and histone modifications, allowing the cell to respond to environmental stimuli and modulate synaptic plasticity.
Epigenetic Regulation of Synaptic Genes: Many genes involved in synapse formation, function, and elimination are under the control of epigenetic regulation. For instance, genes encoding cell adhesion molecules, neurotransmitter receptors, and other synaptic proteins can be epigenetically modulated to fine-tune the establishment and maintenance of synapses.

Epigenetic regulatory mechanisms are critical for directing neuronal pruning and synaptogenesis by modulating gene expression and influencing the molecular processes underlying neural connectivity. These mechanisms allow the nervous system to adapt to experiences, shape neural circuits, and optimize synaptic connections for proper functionality.

Are there specific signaling pathways that are indispensable for the orchestration of neuronal pruning and synaptogenesis?

Yes, several signaling pathways are crucial for the proper orchestration of neuronal pruning and synaptogenesis. These pathways transmit molecular signals that guide the formation, strengthening, and elimination of synapses, as well as the refinement of neural connections. Here are some of the indispensable signaling pathways involved:

Brain-Derived Neurotrophic Factor (BDNF) Pathway: BDNF, a member of the neurotrophin family, is critical for promoting neuronal survival, differentiation, and synaptic plasticity. BDNF signaling through its receptor, TrkB, plays a pivotal role in synaptogenesis and synaptic refinement by enhancing the growth and maintenance of synapses.
Wnt Signaling Pathway: The Wnt pathway is involved in a variety of developmental processes, including neuronal connectivity. Wnt signaling influences axon guidance, dendrite development, and synapse formation by regulating the cytoskeleton and intracellular pathways within neurons.
Notch Signaling Pathway: The Notch pathway is essential for cell-cell communication and has roles in neural development. Notch signaling influences the balance between neuronal differentiation and maintenance of precursor cells. Disruption of Notch signaling can impact synaptic connectivity.
Ephrin Receptor Pathway: Ephrin receptors and their ligands, ephrins, are involved in axon guidance and synaptic organization. The interaction between ephrins on one neuron and their corresponding receptors on another plays a role in shaping synaptic connections and neural circuits.
Neuregulin-ErbB Pathway: Neuregulins, ligands that activate ErbB receptor tyrosine kinases, are involved in the development of glial cells and synapses. This pathway plays a role in coordinating the formation of pre- and postsynaptic elements during synaptogenesis.
Calcium Signaling: Intracellular calcium plays a critical role in neuronal activity and synaptic plasticity. Calcium signaling is involved in synaptic vesicle release, postsynaptic response, and the activation of various signaling cascades that influence synaptogenesis.
Activity-Dependent Pathways: Neuronal activity itself, often initiated by synaptic transmission, triggers signaling pathways that contribute to synaptic plasticity and connectivity refinement. NMDA receptor-dependent calcium influx is a key player in activity-dependent processes.

Specific signaling pathways are indispensable for directing neuronal pruning and synaptogenesis. These pathways orchestrate various aspects of neural development and connectivity, ensuring the precise formation, strengthening, and elimination of synapses that are essential for the functional wiring of the nervous system.

What regulatory codes maintain and oversee the operation of neuronal pruning and synaptogenesis?

The "regulatory codes" that maintain and oversee the operation of neuronal pruning and synaptogenesis involve a complex interplay of molecular mechanisms, gene expression, and cellular signaling. These codes ensure the precise execution of these processes while adapting to developmental needs and environmental cues. Here are some of the key regulatory elements that govern neuronal pruning and synaptogenesis:

Activity-Dependent Regulation: Neuronal activity, driven by synaptic transmission and sensory experiences, acts as a regulatory code. It guides the strengthening of active synapses and the elimination of less active ones, contributing to the refinement of neural circuits.
Transcriptional Regulation: Transcription factors and other regulatory molecules control gene expression patterns during neuronal development. These factors determine which genes are turned on or off, influencing synaptogenesis, dendritic branching, and other processes.
Epigenetic Modification Patterns: Epigenetic marks, such as DNA methylation and histone modifications, form regulatory codes that impact gene expression. These marks can be dynamically altered in response to neural activity, experience, and environmental factors.
Molecular Signaling Networks: Signaling pathways, such as BDNF-TrkB, Wnt, and Notch, form interconnected networks that convey instructions for synaptogenesis and pruning. These pathways regulate cellular responses to molecular cues.
Neurotrophins and Growth Factors: Neurotrophic factors, like BDNF, NGF, and others, play crucial roles in regulating neuronal survival, differentiation, and synaptic plasticity. They ensure that proper connections are established and maintained.
Guidance Molecules and Receptors: Guidance cues and their receptors direct axon pathfinding and dendritic arborization. These molecules ensure that neurons connect to their correct targets during development.
Cell Adhesion Molecules: Cell adhesion molecules ensure that synapses are formed between appropriate pre- and postsynaptic partners. They contribute to the precise wiring of neural circuits.
MicroRNAs and Non-Coding RNAs: MicroRNAs and other non-coding RNAs regulate gene expression post-transcriptionally. They fine-tune the levels of specific proteins involved in synaptogenesis and pruning.

The regulatory codes governing neuronal pruning and synaptogenesis encompass a diverse array of mechanisms that interact to ensure the proper development and refinement of neural connections. These codes integrate genetic information, molecular signaling, cellular responses, and environmental inputs to sculpt the intricate connectivity of the nervous system.

Is there scientific evidence that supports the notion that neuronal pruning and synaptogenesis were brought about by the process of evolution?

The intricate processes of neuronal pruning and synaptogenesis, fundamental to the development and functionality of the nervous system, present significant challenges for an evolutionary explanation. The idea that these processes could have emerged gradually, through a stepwise evolutionary progression, faces substantial hurdles given the interdependence of various codes, languages, signaling networks, and proteins that must be operational from the beginning. 

Complexity and Functional Requirements: Neuronal pruning and synaptogenesis involve a remarkable level of complexity, requiring precise coordination of multiple molecular interactions and genetic regulations. The establishment of synaptic connections necessitates intricate guidance cues, molecular signaling, and precise cellular interactions. A stepwise evolutionary approach would demand the gradual development of each of these components, without the guarantee of functionality at intermediate stages. It is difficult to conceive how partially formed systems with no immediate function could have been selected for, as they would provide no selective advantage to an organism.
Interdependence and Instantiation: What makes the evolutionary pathway even more implausible is the interdependence of the various components. Signaling pathways, gene expression networks, and molecular codes are not independent entities; they rely on each other for their functionality. A fully operational system is required for neuronal pruning and synaptogenesis to occur. The language of molecular signaling pathways needs a coherent molecular vocabulary that includes proteins, receptors, and other elements. Without all these components functioning together, no functional outcome would be achieved, rendering any intermediate stages non-adaptive and non-selectable.
Coordinated Emergence of Multiple Mechanisms: The coordinated emergence of gene expression regulations, epigenetic modifications, molecular signaling, and cellular interactions is a substantial challenge for stepwise evolution. The likelihood of these mechanisms independently evolving, and then coincidentally aligning to support neuronal pruning and synaptogenesis, stretches the bounds of probability. This complex coordination is best explained by the concept of intelligent design, where all necessary components are instantiated simultaneously to achieve a functional outcome.
Irreducible Complexity and Intelligent Design: The concept of irreducible complexity arises when a system relies on multiple interacting components, none of which can be removed without disrupting function. Neuronal pruning and synaptogenesis could be seen as irreducibly complex systems. These systems were most likely designed and implemented all at once with all their intricate interdependencies in place, rather than evolving in a piecemeal fashion.

The simultaneous emergence of neuronal pruning and synaptogenesis as fully operational systems seems more plausible than the stepwise evolution of their various components. The interdependence of codes, languages, signaling networks, and proteins required for their function, along with the complexity and functional demands of these processes, presents a compelling case for intelligent design as the best explanation for the origins of these intricate mechanisms.

How might the systems and structures involved in neuronal pruning and synaptogenesis be considered irreducibly complex or interdependent?

Neuronal pruning and synaptogenesis represent intricate processes that exhibit features of irreducible complexity and interdependence, reinforcing the notion that they are the result of intelligent design rather than gradual evolution.

Molecular Signaling and Receptors: The language of molecular signaling involves complex interactions between signaling molecules and their receptors. This signaling is indispensable for guiding axons, dendrites, and synaptic connections. The absence of any key signaling component would lead to an incomplete and non-functional process.
Guidance Cues and Cell Adhesion Molecules: The guidance cues that direct the growth of axons and dendrites are interdependent with cell adhesion molecules that enable synaptic connections. Without proper guidance cues, neurons might not reach their targets, and without functional adhesion molecules, synapses would not form properly.
Gene Expression and Transcription Factors: The genetic code, transcription factors, and gene expression are intricately involved in shaping neuronal connectivity. The absence of specific genes or regulatory elements would disrupt the orchestration of synaptogenesis and pruning.
Synaptic Activity and Plasticity: The plasticity of synapses, allowing them to strengthen or weaken based on activity, is intertwined with the overall process of pruning. The absence of synaptic activity would hinder both the refinement of synapses and the elimination of excess connections.
Molecular Codes and Signaling Pathways: The "codes" for molecular signaling pathways must be present alongside functional receptors, ligands, and downstream effectors. The absence of any of these components would result in disrupted communication and misdirection of neural growth.
Epigenetic Regulation and Genetic Expression: Epigenetic modifications, such as DNA methylation and histone modifications, regulate gene expression critical for proper development. The intricate interplay between epigenetic marks, gene expression, and neural connectivity is essential for the successful operation of these processes.

Once neuronal pruning and synaptogenesis are fully instantiated and operational, with which other intra and extracellular systems do they closely interact or rely?

Neuronal pruning and synaptogenesis, once fully instantiated and operational, closely interact with various intra and extracellular systems to ensure the proper development and function of the nervous system. These interactions contribute to the establishment of functional neural circuits and the precise wiring of the brain.

Intracellular Interactions

Intracellular Signaling Pathways: Neuronal pruning and synaptogenesis rely on intricate intracellular signaling pathways that regulate processes such as gene expression, cytoskeletal dynamics, and organelle transport. These pathways help neurons respond to extracellular cues and adapt their connectivity.
Cytoskeletal Dynamics: The cytoskeleton, comprising microtubules, microfilaments, and intermediate filaments, plays a vital role in axon and dendrite growth, guidance, and synaptic plasticity. Cytoskeletal elements are crucial for maintaining neuronal structure and connectivity.
Intracellular Transport Systems: Molecular motors and transport mechanisms facilitate the movement of organelles, vesicles, and other cellular components within neurons. Proper intracellular transport is necessary for the delivery of essential materials to growing axons and dendrites.

Extracellular Interactions:

Synaptic Activity and Neurotransmission: Neuronal pruning and synaptogenesis closely interact with synaptic activity and neurotransmission. Active synapses strengthen through activity-dependent mechanisms, while less active synapses are eliminated through pruning. This interaction fine-tunes neural connectivity.
Cell-Cell Communication: Interactions between neurons and other cell types, such as glia, are crucial for guiding axon growth, providing trophic support, and modulating synaptic connections. These interactions help create a conducive environment for proper neuronal development.
Neurotrophic Factors and Growth Factors: Neurotrophic factors play a key role in promoting neuronal survival, differentiation, and synaptic plasticity. They interact with neuronal pruning and synaptogenesis by influencing cell survival, axon guidance, and synapse formation.
Extracellular Matrix (ECM): The ECM provides physical and molecular cues that guide axon and dendrite growth, influence synaptic maturation, and help establish proper neural circuits. The interactions between neurons and the ECM play a role in shaping neural connectivity.

Epigenetic Regulations and Feedback Loops

Epigenetic Mechanisms: Epigenetic regulations, including DNA methylation and histone modifications, influence gene expression patterns that impact neuronal connectivity. These mechanisms are influenced by neuronal activity, shaping the interactions between synaptic activity and gene expression.
Activity-Dependent Feedback Loops: Synaptic activity influences epigenetic modifications, which in turn affect gene expression. This creates feedback loops that allow the nervous system to adapt to experiences and optimize neural circuitry.

Neuronal pruning and synaptogenesis closely interact with a network of intra and extracellular systems to ensure the proper development, refinement, and maintenance of neural connectivity. These interactions highlight the intricate coordination required for the formation of functional neural circuits and emphasize the complexity of the mechanisms involved in shaping the brain's intricate wiring.

1. Neuronal pruning and synaptogenesis rely on intricate semiotic codes and languages for proper communication and coordination between cells and molecules.
2. The systems involved in neuronal pruning and synaptogenesis are highly interdependent, requiring the simultaneous presence and precise coordination of multiple components for functionality.
3. Interlocking codes and interdependence suggest a carefully designed setup rather than an unguided, stepwise evolutionary process.

1. Wikipedia: Synapse pruning

https://reasonandscience.catsboard.com

8Evolution of the brain Empty Re: Evolution of the brain Fri Sep 01, 2023 4:01 am

Otangelo


Admin

Neurulation and Neural Tube Formation

How do neurulation and neural tube formation provide the foundation for the development of the central nervous system in vertebrates?

Neurulation and neural tube formation are critical processes in vertebrate embryonic development that lay the foundation for the creation of the central nervous system (CNS). These processes intricately shape and transform the embryonic tissue, setting the stage for the formation of the brain and spinal cord.

Neurulation

Neurulation is the initial step in the formation of the CNS. It begins with the transformation of the neural plate, a flat sheet of ectodermal tissue, into the neural tube. This transformative process involves several key stages:

Elevation of Neural Folds: As the embryo develops, the neural plate undergoes a process of elevation, forming neural folds on both sides. These folds gradually approach each other along the midline.
Fusion of Neural Folds: The neural folds eventually fuse at the midline, creating a neural tube. This tube becomes the precursor to the brain and spinal cord.
Formation of Neural Crest Cells: Alongside the neural tube formation, a population of cells known as neural crest cells emerge at the borders of the neural plate. These cells play a crucial role in forming various structures, including peripheral nerves, ganglia, and some skeletal elements.

Neural Tube Formation

The neural tube, formed through neurulation, is the rudimentary structure that gives rise to the brain and spinal cord. It undergoes further specialization to create distinct regions of the CNS:

Primary Vesicle Formation: The neural tube initially differentiates into three primary vesicles: the prosencephalon (forebrain), mesencephalon (midbrain), and rhombencephalon (hindbrain).
Secondary Vesicle Formation: These primary vesicles subsequently undergo further differentiation into five secondary vesicles: telencephalon and diencephalon from the prosencephalon, mesencephalon remains unchanged, and metencephalon and myelencephalon from the rhombencephalon.
Cavities and Structure Formation: These vesicles expand and develop specific cavities that become the ventricles of the brain and central canal of the spinal cord. The walls of these vesicles differentiate into the various regions of the CNS.

Neurulation and neural tube formation are critical because they set the foundation for the complex structures and functions of the CNS. These processes ensure the proper development of the brain and spinal cord, which are essential for sensory perception, motor control, cognition, and a myriad of other neurological functions.

How does the neural tube differentiate into distinct regions, such as the brain and spinal cord?

The neural tube, formed through the process of neurulation, gives rise to both the brain and the spinal cord in vertebrate embryos. This remarkable differentiation involves complex molecular signaling and patterning mechanisms that lead to the formation of distinct regions with specific functions.

Formation of Primary Vesicles

After the initial fusion of the neural folds, the neural tube differentiates into three primary vesicles along the anterior-posterior axis: the prosencephalon (forebrain), mesencephalon (midbrain), and rhombencephalon (hindbrain). Each primary vesicle serves as the basis for further differentiation.

Secondary Vesicle Formation and Patterning

These primary vesicles then undergo further differentiation into five secondary vesicles through a process called regionalization:

Telencephalon and Diencephalon (Forebrain): The prosencephalon gives rise to the telencephalon (which develops into the cerebral hemispheres) and the diencephalon (which forms structures like the thalamus and hypothalamus).
Mesencephalon (Midbrain): The mesencephalon remains relatively unchanged and develops into the midbrain structures, including the tectum and tegmentum.
Metencephalon and Myelencephalon (Hindbrain): The rhombencephalon differentiates into the metencephalon (developing into the pons and cerebellum) and the myelencephalon (forming the medulla oblongata).

Patterning Signals and Genetic Regulation

The differentiation of the neural tube into these distinct regions is governed by intricate molecular signaling pathways, including the actions of morphogens such as Sonic Hedgehog (Shh), Fibroblast Growth Factors (FGFs), and Bone Morphogenetic Proteins (BMPs). These signaling molecules establish concentration gradients along the neural tube, instructing cells to adopt specific identities based on their location.

Hox Genes

Hox genes, which play a pivotal role in determining regional identities along the anterior-posterior axis of the body, are also crucial for neural tube differentiation. The expression patterns of Hox genes guide the formation of different segments within the neural tube.

Cellular Migration and Differentiation

As cells within the neural tube receive specific signaling cues, they migrate to their designated regions and differentiate into the diverse cell types that make up the brain and spinal cord.

Patterning and Function

This complex differentiation process ultimately gives rise to the various brain structures and spinal cord segments, each with specialized functions that contribute to sensory perception, motor control, cognition, and other essential neurological processes.

Evolution of the brain 2912_n10
Transverse sections that show the progression of the neural plate to the neural groove from bottom to top 1

At what juncture in the evolutionary timeline are neurulation and neural tube formation postulated to have made their first appearance?

Neurulation and neural tube formation are fundamental embryological processes that lead to the development of the central nervous system (CNS), including the brain and spinal cord. These events are critical for the formation of complex nervous systems and have been conserved across a broad range of vertebrates. Here's a look into their possible evolutionary origins:

Origins of the Nervous System

Simple Nervous Systems: The earliest multicellular organisms would have had rudimentary nervous systems, consisting of simple nerve nets or basic nerve cords. These basic nervous structures wouldn't have required specialized processes like neurulation.
Bilateria and CNS Development: The appearance of bilaterally symmetrical animals, or Bilateria, is a key event in the evolution of the CNS. It is hypothesized that the ancestors of modern bilaterians possessed a centralized nerve cord, which served as a precursor to more advanced nervous systems.
Neurulation and Neural Tube Formation: Neurulation and the formation of a neural tube would have emerged with the need for a more centralized and organized nervous system. This process would have been critical for the development of a dorsal nerve cord in early chordates, which is an ancestral feature of all vertebrates.

Vertebrate Evolution and Neural Tube Specialization

Primitive Chordates: In early chordates like amphioxus, a simple notochord and nerve cord were present. These organisms would have exhibited basic neurulation processes, leading to the formation of a dorsal nerve cord.
Early Vertebrates: With the emergence of early vertebrates, the neural tube would have become more specialized, giving rise to distinct regions such as the forebrain, midbrain, and hindbrain. This differentiation is crucial for the diverse functions and capabilities seen in modern vertebrates.
Neural Crest Cells: Along with the neural tube, the evolution of neural crest cells would have played a pivotal role in vertebrate diversification. These cells originate from the borders of the neural tube and migrate to various parts of the embryo, contributing to structures like cranial bones, peripheral nerves, and more.

The processes of neurulation and neural tube formation are thought to have made their appearance during the evolution of early chordates, setting the stage for the complex nervous systems seen in today's vertebrates. These developmental events would have provided the architectural foundation for advanced neural structures, facilitating sophisticated behaviors and adaptations in vertebrate lineages.

Which de novo genetic information would be requisite to instantiate the processes of neurulation and neural tube formation?

Neurulation and neural tube formation are critical stages in the development of the vertebrate central nervous system. They rely on intricate molecular and cellular processes that are guided by various genes and their corresponding proteins. While numerous genes are involved in this complex developmental process, certain genes are recognized as core players in driving neurulation and establishing the neural tube. Here's an overview of some of these crucial genetic components:

Key Genetic Components

Notochord Induction Genes: The notochord, a midline embryonic structure, secretes signaling molecules that instruct the overlying ectoderm to become neural tissue. Genes like Noggin, Chordin, and Follistatin are crucial for this induction, as they inhibit proteins that would otherwise prevent neural differentiation.
Neural Plate Border Specifiers: Genes such as Pax3, Pax7, Msx1, and Zic1 play roles in specifying cells at the border of the neural plate. These border cells can give rise to both neural crest cells and neural tissue.
Neural Fold Elevation and Convergence: As the neural plate forms, it starts to fold, with its edges (neural folds) elevating and moving towards each other. Genes like Shh (Sonic hedgehog) and BMP4 (Bone Morphogenetic Protein 4) play roles in guiding this morphogenesis.
Neural Tube Closure: The eventual fusion of the neural folds to form a closed neural tube is a critical step. Genes such as Celsr1, Vangl2, and Fzd3 are vital components of the planar cell polarity pathway and are instrumental in coordinating the movements of cells during tube closure.
Neural Differentiation and Patterning: Once the neural tube is formed, it undergoes further differentiation and patterning. Genes like Shh and Wnt are involved in ventral and dorsal patterning of the neural tube, respectively, establishing regions that will later give rise to different structures in the CNS.

The process of neurulation and neural tube formation is orchestrated by a myriad of genes working in concert. These genes provide the de novo genetic information necessary for the successful development of the central nervous system. Any disruptions in the function of these genes can lead to neural tube defects, highlighting their critical importance in embryonic development.

What specific manufacturing codes and languages would have to emerge and be operational for neurulation and the formation of the neural tube?

Neurulation and the formation of the neural tube are intricate processes in embryonic development, driven by a series of tightly regulated molecular and cellular instructions. To understand these "manufacturing codes and languages," one must delve into the complex world of genetic regulation, signaling pathways, and cell-to-cell communications that drive these developmental processes. Here's a glimpse into some of these genetic "codes" and "languages":

Genetic Codes and Regulation

Gene Expression and Transcription Factors: Specific genes are turned on or off during different stages of neurulation. Transcription factors like Sox1, Sox2, and Sox3 are expressed in the early neural plate and are crucial for neural differentiation.
Epigenetic Regulation: Modifications to DNA and its associated proteins can alter gene expression without changing the underlying DNA sequence. Epigenetic changes, such as DNA methylation or histone modifications, are pivotal in determining cell fate during neural tube formation.

Signaling Pathways

Bone Morphogenetic Proteins (BMPs) and Their Antagonists: BMP signaling tends to promote epidermal fates, while its inhibition by molecules like Noggin, Chordin, and Follistatin promotes neural fates.
Sonic Hedgehog (Shh) Signaling: The notochord produces Shh, which plays a crucial role in ventral patterning of the neural tube, determining different neuronal subtypes based on concentration gradients.
Wnt Signaling: Important for dorsal patterning of the neural tube and interacts with other signaling pathways to ensure the right balance of cell types.

Cellular Communication and Interaction

Planar Cell Polarity (PCP) Pathway: This pathway controls the convergent extension movements during neurulation, where cells intercalate and the neural plate narrows and lengthens. Key components include Vangl2, Celsr1, and Fzd3.
Cell Adhesion Molecules: Molecules such as cadherins and integrins play roles in ensuring that cells stick together and move collectively during the bending and folding processes of neurulation.

Neurulation and neural tube formation are orchestrated by a myriad of "manufacturing codes and languages" at the genetic, molecular, and cellular levels. These intricate processes ensure the proper development and functionality of the central nervous system. Any disruptions in these instructions can lead to neural tube defects, emphasizing their vital importance in embryonic development.

Which epigenetic regulatory mechanisms are pivotal in guiding the processes of neurulation and neural tube formation?

Neurulation and neural tube formation are intricate events during embryonic development. These processes are not solely governed by the genomic DNA sequence but also by epigenetic modifications that influence gene expression. Epigenetics, meaning "above genetics," involves chemical modifications to DNA and histones, non-coding RNAs, and chromatin remodeling, which collectively shape the way genes are expressed. Here's a look into some of the epigenetic regulatory mechanisms crucial for neurulation and neural tube formation:

DNA Methylation

DNA Methyltransferases (DNMTs): These enzymes add methyl groups to the cytosine residues in DNA, typically leading to gene silencing. DNMTs play vital roles in neural differentiation and neural tube formation. Anomalous methylation patterns can disrupt the expression of genes essential for these processes.

Histone Modifications

Histone Acetylation and Deacetylation: Acetylation, typically associated with gene activation, is governed by histone acetyltransferases (HATs). In contrast, deacetylation, linked with gene repression, is controlled by histone deacetylases (HDACs). These modifications are crucial in determining the transcriptional activity of genes involved in neurulation.
Histone Methylation: Depending on the specific lysine residue modified and the number of added methyl groups, histone methylation can either activate or repress gene expression. Enzymes like histone methyltransferases and demethylases regulate these modifications, ensuring proper gene expression during neural development.

Chromatin Remodeling

SWI/SNF Complex: This multi-protein complex changes the position of nucleosomes on DNA, allowing or hindering the binding of transcriptional machinery to DNA. This remodeling is essential for the timely activation and repression of genes during neural tube formation.

Non-Coding RNAs

MicroRNAs (miRNAs): These short RNA molecules do not code for proteins but play significant roles in post-transcriptional gene regulation. By targeting specific messenger RNAs (mRNAs), miRNAs can inhibit their translation or lead to their degradation, thus controlling the levels of proteins essential for neurulation.
Long Non-Coding RNAs (lncRNAs): These RNA molecules, longer than miRNAs, can interact with DNA, RNA, or proteins. They play roles in various cellular processes, including the regulation of gene expression at both transcriptional and post-transcriptional levels during neural development.

The orchestration of neurulation and neural tube formation is an intricate ballet of gene expression, with epigenetic regulatory mechanisms serving as the choreographers. Proper epigenetic modifications ensure that the right genes are expressed at the right time, facilitating the harmonious development of the neural tube and, subsequently, the central nervous system.

Are there distinct signaling pathways that are essential for the seamless orchestration of neurulation and neural tube formation?

Neurulation and neural tube formation are complex processes that require precise coordination of cellular behavior. For this to occur, multiple signaling pathways operate in tandem, dictating cell fate, proliferation, migration, and morphogenesis. The following pathways have been recognized as pivotal in guiding the processes of neurulation and neural tube formation:

Sonic Hedgehog (Shh) Signaling

Dorsal-Ventral Patterning: Shh, secreted by the notochord and floor plate, is instrumental in the ventral patterning of the neural tube. It specifies the identity of ventral neural cell types by inducing various transcription factors.

Bone Morphogenetic Protein (BMP) Signaling

Neural Induction: BMPs, members of the TGF-β superfamily, play a critical role in ectodermal patterning. BMP antagonists, secreted by the organizer tissues, such as noggin, chordin, and follistatin, promote neural induction by inhibiting BMP activity.

Wnt Signaling

Neural Plate Border and Neural Crest Specification: Wnt signaling pathways, particularly canonical Wnt/β-catenin signaling, have pivotal roles in specifying the neural plate border and inducing the neural crest, a population of cells that gives rise to a plethora of derivatives, including peripheral neurons and glial cells.

Retinoic Acid (RA) Signaling

Anterior-Posterior Patterning: RA, a derivative of Vitamin A, produced in the posterior neural tissue, helps in establishing anterior-posterior identities within the neural tube. It operates in gradient fashion, with higher concentrations leading to more posterior neural fates.

Fibroblast Growth Factor (FGF) Signaling

Neural Induction and Patterning: FGFs have diverse roles during neurulation, including promoting neural induction and aiding in patterning the neural plate by working alongside other signaling pathways.

Planar Cell Polarity (PCP) Signaling

Convergent Extension Movements: PCP signaling is crucial for the cellular movements that shape the neural plate and tube. Convergent extension movements, driven by this pathway, elongate the neural plate along the anterior-posterior axis and narrow it mediolaterally.

The orchestration of neurulation and neural tube formation hinges on a symphony of signaling pathways that work in harmony. These pathways, sensitive to gradients and timing, collectively guide the cellular behaviors and fate decisions necessary for the construction of a well-formed neural tube, the precursor to the central nervous system.

What are the regulatory codes that underpin and oversee the mechanisms of neurulation and neural tube formation?

Neurulation and the formation of the neural tube are foundational processes during vertebrate embryogenesis that give rise to the central nervous system. These processes are underpinned by a complex interplay of molecular, cellular, and mechanical codes that ensure their proper execution. The following regulatory codes are central to the oversight and execution of these processes:

Gene Regulatory Networks (GRNs)

Master Regulators: Transcription factors such as Sox2, Pax3, and Pax7 are pivotal in initiating and maintaining neural identity during the early stages of neural plate formation. These regulators initiate gene cascades crucial for successive phases of neurulation.
Coordinating Morphogenesis: Certain genes ensure the proper bending, folding, and closure of the neural plate. For instance, genes coding for cell-adhesion molecules like N-cadherin help in maintaining tissue integrity during these morphogenetic movements.

MicroRNAs (miRNAs)

Post-transcriptional Regulation: miRNAs, small non-coding RNAs, modulate gene expression post-transcriptionally. They're involved in fine-tuning the dynamics of protein production necessary for neural tube formation. For example, miR-34 and miR-449 have been implicated in regulating neural crest cell migration and differentiation.

Epigenetic Modifications

Histone Modifications and DNA Methylation: Chemical modifications to DNA and histones, like methylation and acetylation, modulate the accessibility of genes to the transcriptional machinery, thus influencing gene expression patterns during neurulation.

Feedback Loops

Ensuring Robustness: Many of the signaling pathways, such as Shh and BMP, involved in neural tube formation have built-in feedback loops. These loops help ensure that the processes are robust against perturbations and are carried out with fidelity.

Mechanical Forces

Cell Shape and Tissue Morphogenesis: Cellular behaviors, such as apical constriction and cell intercalation, are driven by mechanical forces. These behaviors, in turn, drive the neural plate's bending and folding. Regulatory codes, often in the form of mechanotransduction pathways, ensure that these forces are generated and applied correctly.

The precise orchestration of neurulation and neural tube formation relies on a comprehensive set of regulatory codes, ranging from gene expression and post-transcriptional modifications to mechanical forces. Together, these codes ensure that the embryo develops a well-formed neural tube, setting the stage for the later development of the brain and spinal cord.

Does current scientific literature provide evidence to suggest that neurulation and neural tube formation were evolutionary processes?

Neurulation and neural tube formation are foundational processes during vertebrate embryogenesis that give rise to the central nervous system. These processes involve a myriad of intricate and interdependent molecular, cellular, and mechanical mechanisms, suggesting the complexity of the design and the challenges of evolutionary explanations.

Complexity and Interdependence

Integrated Gene Regulatory Networks (GRNs): Neurulation is driven by a complex set of GRNs that not only need to be present but also intricately tuned to ensure proper timing and patterning of neural development. The coordinated action of master regulators, like Sox2, Pax3, and Pax7, is pivotal. Any disruption or incomplete integration of these GRNs would likely result in non-functional or adverse outcomes.
Signaling Pathways: Key pathways such as Sonic Hedgehog (Shh) and Bone Morphogenic Protein (BMP) have tightly integrated feedback loops ensuring that neural plate cells receive the right signals at the right time. Without the full signaling pathway present and functional, the entire process could be derailed.
Epigenetic Controls: DNA methylation, histone modifications, and other epigenetic controls are necessary for precise temporal and spatial gene expression during neurulation. These controls are not just add-ons but essential layers of regulation.

Challenges for Stepwise Evolution

Coordinated Cellular Behaviors: The physical act of neurulation, where the neural plate bends, folds, and eventually fuses to form the neural tube, requires a multitude of cells to act in concert. These behaviors, driven by mechanical forces and cellular signaling, seem to necessitate a pre-existing set of instructions rather than a gradual, stepwise accumulation.
Symbiotic Protein Interactions: Many proteins involved in neurulation interact in ways that seem symbiotic. For instance, cell adhesion molecules ensure tissue integrity during the folding of the neural plate. The presence of one protein without its partner or counterpart might not only be non-functional but could be detrimental.

The Problem of Intermediates

Functionality of Partial Systems: For evolution to favor a particular trait or mechanism, it generally needs to confer some advantage. However, with neurulation, it's challenging to envision how partial or intermediate stages could offer any functional advantage. Incomplete neural tube formation results in severe abnormalities.
Requirement for Simultaneous Systems: The codes, languages, signaling, and proteins involved in neurulation seem to be so interdependent that they must all be in place for the process to work. The idea of them evolving simultaneously, yet independently, stretches the imagination.

While the scientific community continues to explore the mechanisms and origins of complex processes like neurulation, the sheer intricacy, and interdependence of the involved systems raise profound questions about the feasibility of stepwise evolutionary explanations. The presence of such a well-coordinated and integrated system suggests a design of profound intelligence.

Could the mechanisms and components involved in neurulation and neural tube formation be characterized as irreducibly complex or interdependent?

Neurulation and neural tube formation represent quintessential processes that give rise to the central nervous system in vertebrate embryogenesis. The assembly and function of the structures and pathways within this framework seem to present a deeply interdependent and potentially irreducibly complex system.

Irreducible Complexity and Interdependence

Gene Regulatory Networks (GRNs): Neurulation is underpinned by an intricate set of GRNs, where master regulatory genes like Sox2, Pax3, and Pax7 are pivotal. A failure in one aspect of this network could compromise the entire process. These genes and their networks function collectively, with one component being non-functional in the absence of the others.
Signaling Pathways: Key pathways, including Sonic Hedgehog (Shh) and Bone Morphogenic Protein (BMP), are not just sequences of events, but possess tightly integrated feedback mechanisms. If one part of these pathways was missing or non-functional, it could jeopardize the entire process of neural differentiation.
Cellular Mechanisms and Dynamics: The cellular behaviors during neurulation, from cell migration to changes in cell shape and polarity, hinge on a balance of forces and cellular communications. The mechanisms driving these behaviors seem interdependent, as a malfunction in one would impair the entire physical process of neurulation.
Epigenetic Regulation: DNA methylation, histone modifications, and non-coding RNAs contribute to precise gene expression during neurulation. These components form an interconnected regulatory system, where the absence or malfunction of one aspect could lead to catastrophic developmental errors.

The Cross-Talk and Communication Systems

Intercellular Communication: Cells during neurulation do not operate in isolation. They communicate using signaling molecules, such as growth factors, to ensure synchronized behavior. This communication is vital for the seamless orchestration of cell movements and differentiation.
Intracellular Communication: Within each cell, multiple pathways and molecular processes, from protein synthesis to cellular metabolism, are interconnected. Proteins, metabolites, and ions continually communicate, ensuring the cell's function and survival.

The Evolutionary Implications

Challenge of Stepwise Evolution: Given the myriad of codes, languages, signaling pathways, and proteins involved in neurulation, the evolutionary progression of such an intricate system in a stepwise manner becomes daunting. Intermediate stages might not provide any functional advantage, making natural selection of such stages implausible.
Requirement for Simultaneous Systems: The sheer interdependence means that for one system to function properly, others must already be in place. It challenges the notion of gradual addition, as adding one component without the others could result in a non-functional or even detrimental system.

The profound complexity and interdependence observed in neurulation and neural tube formation are awe-inspiring. Such intricately connected systems, where the absence of one component could lead to the collapse of the entire process, suggest a sophisticated design that goes beyond the capabilities of random, stepwise evolutionary processes.

Once neurulation and neural tube formation are fully operational, what other intra and extracellular systems might they be intricately interconnected with or dependent upon?

Once neurulation and neural tube formation processes are fully realized, they don't act in isolation. The neural tube and its constituent cells become an active hub, intricately connected to various other cellular systems and external influences. These connections and dependencies ensure the proper functioning, differentiation, and survival of the neural tissue.

Intracellular Systems

Cellular Metabolism: Neurons, and the glial cells supporting them, have high metabolic demands. The mitochondria, often referred to as the cellular powerhouses, must supply this demand by producing ATP, and their health and function are vital for neural cell survival.
Protein Synthesis and Degradation: Neural cells constantly produce proteins necessary for synaptic function, neurotransmitter synthesis, and cell maintenance. Ribosomes synthesize these proteins, while proteasomes and lysosomes degrade misfolded or old proteins.
Calcium Signaling: Intracellular calcium levels in neurons are critical for processes like neurotransmitter release, gene expression, and synaptic plasticity. The endoplasmic reticulum, mitochondria, and various ion channels coordinate to manage these levels.

Extracellular Systems and Influences

Neurotrophic Factors: These are molecules that support neuronal survival, differentiation, and growth. Molecules such as nerve growth factor (NGF) or brain-derived neurotrophic factor (BDNF) are essential for the health and function of neurons.
Glial Support: Astrocytes, oligodendrocytes, and microglia provide nutritional, structural, and immune support to neurons. They are not merely passive support cells but play active roles in synaptic function, myelination, and neural defense.
Synaptic Communication: Neurons communicate with each other via synapses, where neurotransmitters like glutamate, GABA, or dopamine are released. This neurotransmitter system is paramount for neural communication and information processing.
Vascular Supply: Blood vessels provide essential nutrients and oxygen to the neural tissue. Moreover, the blood-brain barrier, formed by the interaction of endothelial cells, astrocytes, and pericytes, protects the brain from harmful substances while ensuring the supply of necessary nutrients.
Extracellular Matrix (ECM): The ECM provides structural support and plays a role in guiding cell migration during development. It also influences cell behavior, synaptic stability, and plasticity in the mature nervous system.

The completion of neurulation and neural tube formation is just the beginning of a series of intricate relationships and dependencies that neural cells will establish with both internal cellular systems and external influences. This highly integrated network ensures the optimal functionality and adaptability of the central nervous system throughout an organism's life.

1. If complex systems exhibit properties of interdependence, semiotic coding, and synchronization, implying that their elements had to emerge simultaneously and harmoniously to function properly, then such systems show traits commonly attributed to designed mechanisms.
2. The neurulation and neural tube formation processes, along with their associated intracellular and extracellular systems, exhibit these very properties of interdependence, semiotic coding, and synchronization.
3. Therefore, the neurulation and neural tube formation processes, along with their connected systems, indicate traits commonly attributed to designed mechanisms.

1. Wikipedia: Neurulation

https://reasonandscience.catsboard.com

9Evolution of the brain Empty Re: Evolution of the brain Wed Oct 04, 2023 6:12 am

Otangelo


Admin

Brain regions 


Cerebrum: The cerebrum is the largest part of the brain, responsible for higher cognitive functions. It consists of the two cerebral hemispheres and is covered by the cerebral cortex.
Within the cerebrum, we find structures such as:
- Basal Ganglia: A group of nuclei in the brain associated with a variety of functions including control of voluntary motor movements, procedural learning, and cognition.
- Limbic System: A complex set of brain structures responsible for emotion, behavior, motivation, and long-term memory. Within the limbic system, you'll find:
  - Amygdala: Involved in emotion processing and the formation of emotional memories.
  - Hippocampus: Essential for memory formation.
  - Hypothalamus: Regulates numerous bodily functions, including hunger, thirst, body temperature, and circadian rhythms. It also connects the nervous system to the endocrine system via the pituitary gland.

Midbrain: Part of the brainstem, it's associated with vision, hearing, motor control, alertness, and temperature regulation.

Brainstem: Connects the cerebrum and cerebellum to the spinal cord. It plays a critical role in the control of several vital bodily functions, including:
- Breathing
- Digestion
- Heart rate
- Blood pressure

It's worth noting that while some structures like the amygdala and hypothalamus are part of broader categories like the limbic system, they have very distinct and vital roles. The brain is a highly intricate and interconnected organ, and while categorizing helps in understanding, the divisions can sometimes be fluid.

Cerebrum: Divided into two hemispheres, the left and right, it is further segmented into distinct lobes:
   - Frontal Lobe: Important for cognitive skills such as emotional expression, problem-solving, memory, and judgment.
   - Parietal Lobe: Primarily focused on processing sensory information from various parts of the body.
   - Temporal Lobe: Associated with processing auditory information and encoding memory.
   - Occipital Lobe: Dedicated to visual processing.

Limbic System: Often termed the emotional brain, it includes:
   - Amygdala: Processing of emotions and determination of what memories are stored and where.
   - Hippocampus: Essential for learning and memory.
   - Thalamus: Acts as a relay station, transmitting information between the cerebellum and cerebrum.
   - Hypothalamus: Connects the nervous system to the endocrine system via the pituitary gland.

Basal Ganglia: A collection of nuclei that play a central role in movement control, emotion, and cognition.

Brainstem: The posterior part of the brain, continuous with the spinal cord. Within the brainstem, you find:
   - Midbrain: Contains reflex centers associated with eye and head movements.
   - Pons: Bridges the cerebrum with the cerebellum and contains nuclei involved in somatic and visceral motor control.
   - Medulla Oblongata: Regulates several crucial functions including cardiovascular and respiratory systems.

Cerebellum: Positioned under the cerebrum, its main function is coordination of voluntary movements, balance, and posture.

Diencephalon: This encompasses the thalamus and hypothalamus, vital for sensory and motor signal relay and the control of the autonomic functions of the peripheral nervous system.

Corpus Callosum: A large bundle of neural fibers that connect the left and right cerebral hemispheres, allowing for communication between both halves.

Ventricular System: Comprising of four interconnected ventricles, it produces cerebrospinal fluid.

The above are some of the major regions and structures of the human brain, with each having its unique functions and intricacies. The human brain's organization is vast and interconnected, and the divisions and categorizations can be even more detailed based on the context of study.

Celly types in the Human Brain

Neurons: These are the primary electrically excitable cells that process and transmit information.
Glia (or Glial Cells): These cells support and nourish neurons, among other functions.
Vascular Cells: The brain has an extensive vascular network.
Stem and Progenitor Cells: Cells capable of dividing and producing new cells.

Codes operating in the brain

The term "biological codes" in the context of the brain often refers to ways information is encoded and processed in neural systems. The most traditionally recognized code in the brain is the neural code. However, understanding how the brain encodes and processes information is a multifaceted challenge, and there are several proposed mechanisms by which the brain might encode information. Here are some of the primary modes of encoding:

Rate Coding: This involves the rate or frequency of action potentials (or "spikes"). Neurons can vary the frequency of these action potentials to convey different intensities of a stimulus.
Temporal Coding: This concerns the precise timing of action potentials. The idea here is that the exact times at which neurons fire can convey information.
Population Coding: Individual neurons can be noisy and unreliable. However, when the activity of a group of neurons is considered, the combined activity can provide a clearer and more reliable representation of information.
Synchronous Firing: Some theories suggest that neurons that fire simultaneously or in synchrony might be representing the same type of information or be part of the same functional network.
Oscillatory Phase Coding: Information can be encoded in the phase of oscillatory neural activity, particularly in relation to the underlying oscillatory activity of neuronal populations.
Spatial Coding: Involves the location of active neurons in a particular region of the brain. A classic example is the "place cells" in the hippocampus that represent specific locations in space.

Metabolic and Neurochemical Codes: Apart from electrical activity, changes in metabolism and neurotransmitter concentrations also encode information. For instance, neurotransmitters like dopamine can represent reward prediction errors.

Rate, Temporal, and Population Coding: These general encoding mechanisms can be seen as fundamental underpinnings for various specialized codes, such as:
- The Binaural Code: This involves temporal patterns to localize sound.
- The Magnitude Neuronal Codes: Possibly employing rate coding to signify intensity.
- The Memory and Mnemonic Codes: Employing population coding across distributed networks to encode memories.
- The Speech Code: Likely making use of temporal patterns for speech comprehension and production.
- The Visual Code: Utilizing population coding to interpret visual stimuli.
- The Perception Code: Embodying several coding modalities to interpret sensory information.
- The Oscillatory Activity Code: Involving synchronous neuronal firing patterns to facilitate cognitive functions.

Spatial Coding and Synchronous Firing: These are particularly relevant to processes where neuronal position or synchronous activity is pivotal:
- The Axon Guidance Codes: Axon positioning is key for neural development.
- The Cadherin Neuronal Code: Since cadherins ensure proper neuronal adhesion and positioning.
- The Synaptic Code: Relates to the spatial distribution of synapses and their effective communication.

Metabolic and Neurochemical Codes: These link closely with the more molecular and biochemical aspects of neural function:
- The Apoptosis Code: Relating to the biochemistry of cell death.
- The Serotonin Code: As serotonin is a neurotransmitter with distinct chemical properties and functions.
- The Neurotransmitter Code: This involves biochemical interactions of various neurotransmitters.
- The Metabolic Code: Overseeing the essential bioenergetics of the brain.
- The Protein Allosteric, Binding, and Post-translational modification Codes: Managing different aspects of protein interactions and functions.
- The RNA Recognition Code: Concerning molecular interactions affecting RNA processing.
- The Polycomb & Trithorax Codes: Involved in epigenetic regulation of brain functions.

Oscillatory Phase Coding: Can be seen as a specialized type of:
- The Oscillatory Activity Code: Where synchronized neural oscillations play a pivotal role.

The intricate organization of the brain means that many of these codes likely interact and overlap in various ways. The "codes" represent our ongoing effort to understand the multifaceted ways in which the brain encodes, processes, and uses information.




Evolution of the brain Sem_t121

https://reasonandscience.catsboard.com

10Evolution of the brain Empty Re: Evolution of the brain Wed Oct 04, 2023 6:25 am

Otangelo


Admin

List of neuron types


Chandelier Cells (Cerebral Cortex): Inhibitory interneurons that control the activity of pyramidal cells.
Neurogliaform Cells (Cerebral Cortex): Release GABA and regulate local neural circuits.
Double Bouquet Cells (Cerebral Cortex): Inhibitory interneurons involved in columnar processing.
Rosehip Neurons (Human Cortex): Recently discovered inhibitory neurons with a unique morphology and connectivity.
Kisspeptin Neurons (Hypothalamus): Crucial for puberty initiation and fertility.
Orexin (Hypocretin) Neurons (Hypothalamus): Control appetite and wakefulness.
Parvalbumin-positive interneurons (Throughout the Brain): A subclass of GABAergic interneurons involved in rhythm generation.
Cajal-Retzius Cells (Cerebral Cortex): Play a role during cortical development.
Cerebellar Golgi Cells (Cerebellum): Inhibitory interneurons that form synapses with granule cells.
Hippocampal CA1, CA2, CA3 Neurons (Hippocampus): Principal excitatory neurons with distinct connectivity patterns.
Somatostatin-expressing interneurons (Throughout the Brain): Inhibitory neurons that control the activity of excitatory cells.
Vasoactive Intestinal Polypeptide (VIP) Neurons: Modulate the activity of other interneurons.
Intrinsically Photosensitive Retinal Ganglion Cells (ipRGCs) (Retina): Detect light and regulate circadian rhythms.
Arcuate Nucleus Neurons (Hypothalamus): Involved in energy homeostasis and feeding behavior.
Anterior Horn Cells (Spinal Cord): Motor neurons affected in conditions like polio.
Mitral Cells (Olfactory Bulb): Transmit olfactory information to the brain.
Tufted Cells (Olfactory Bulb): Similar to mitral cells, they process olfactory signals.
Müller Cells (Retina): The main glial cells supporting neuronal functions in the retina.
Saccule and Utricle Hair Cells (Inner Ear): Detect linear acceleration and gravity.
Olfactory Sensory Neurons: Detect odors and transmit them to the olfactory bulb.
Cone Bipolar Cells (Retina): Process visual information from cone photoreceptors.
Rod Bipolar Cells (Retina): Process visual signals from rod photoreceptors.
Proprioceptors (Muscles, Tendons): Detect body and limb position.
Neuroendocrine Cells (Hypothalamus): Release hormones directly into the bloodstream.
NG2 Glia (Polydendrocytes) (Throughout the Brain): Progenitor cells that can differentiate into oligodendrocytes.
Cortical Thalamic Relay Neurons: Transmit sensory information from the thalamus to the cortex.
Spiny Projection Neurons (SPNs) (Striatum): Key players in basal ganglia circuits.
Basket Cells (Cerebral Cortex): Inhibitory interneurons that control pyramidal neuron activity.
Ganglionar Eminence-derived interneurons: Migrate during development to populate various brain regions.
Cerebellar Nuclei Neurons: Send output from the cerebellum to other brain areas.
Perineuronal Nets: Specialized extracellular matrix structures that enwrap certain neurons.
Neuroepithelial Cells: Early neural progenitor cells during development.
Radial Glia: Play roles in neurogenesis and guiding migrating neurons.
Ganglion Cells (Peripheral Nervous System): Clusters of neuron cell bodies outside the CNS.
Taste Receptor Cells (Tongue): Detect different taste qualities (sweet, salty, etc.).
Enterochromaffin Cells (Gut): Release serotonin in response to food.
Node of Ranvier: Gaps in the myelin sheath where ion exchange occurs.
Pituicytes (Pituitary): Glial cells supporting the function of the pituitary gland.
Interstitial Cells of Cajal (Gastrointestinal Tract): Act as pacemakers for smooth muscle contraction.
Ia Afferents: Proprioceptive fibers from muscle spindles.
Lamellar (Pacinian) Corpuscles: Detect vibrations and deep pressure.
Tactile (Meissner) Corpuscles: Detect light touch.
Ruffini Endings: Detect skin stretch and torque.
Dorsal Root Ganglion Neurons: Sensory neurons that transmit peripheral signals to the spinal cord.
Pinealocytes (Pineal Gland): Cells that produce melatonin.
Horizontal GABAergic interneurons (Striatum): Regulate output pathways of the basal ganglia.
Suprachiasmatic Nucleus (SCN) Neurons: The primary circadian clock of the brain.
Neurokinin-1 Receptor Neurons: Involved in pain perception.
Locus Coeruleus Noradrenergic Neurons: Major source of brain norepinephrine.
Preganglionic Autonomic Neurons: Neurons that synapse onto autonomic ganglia.
Postganglionic Autonomic Neurons: Receive input from preganglionic neurons and innervate target tissues.
Lewy Bodies: Abnormal protein aggregates found in dopaminergic neurons in Parkinson's Disease.
Area Postrema Neurons: A brain area without a blood-brain barrier, sensitive to circulating toxins.
Edinger-Westphal Nucleus Neurons: Involved in pupillary reflex and lens control.
Magnocellular Neurosecretory Cells (Hypothalamus): Produce oxytocin and vasopressin.
End Bulbs of Krause: Sensory receptors sensitive to cold.
Cortical Column: A functional unit of the cerebral cortex.
Ventricular Zone Cells: Area where neurogenesis primarily occurs during development.
Subventricular Zone Cells: Secondary site of neurogenesis in the developing and adult brain.
Eosinophilic Granule Cells (Pituitary): Release hormones.
Carotid Body Cells: Chemoreceptors that detect changes in blood pH and oxygen.
Brodmann Areas: Regions of the cortex defined by their cytoarchitecture.
Corticothalamic Neurons: Send feedback signals from the cortex to the thalamus.
Nucleus Accumbens Neurons: Involved in reward and reinforcement.
Habenula Neurons: Convey signals from the forebrain to the midbrain.
Nucleus Basalis Neurons: Major source of cholinergic projections to the cortex.
Outer Hair Cells (Inner Ear): Amplify sound-induced vibrations.
Supporting Cells (Inner Ear): Provide structural and functional support to sensory hair cells.
Tan Cells (Spinal Cord): Function in the processing of sensory input.
Zona Incerta Neurons: Involved in sensory-motor integration.
Parabrachial Nucleus Neurons: Process sensory information related to taste and pain.
Kolliker-Fuse Nucleus Neurons: Involved in respiratory rhythm generation.
Medial Geniculate Body Neurons: Process auditory information.
Lateral Geniculate Body Neurons: Process visual information.
Islands of Calleja Neurons: Tiny clusters of granule cells in the ventral striatum.
Anterior Insula Neurons: Process interoceptive and emotional information.
Septal Nucleus Neurons: Involved in reward and reinforcement.
Neurofibrillary Tangles: Protein aggregates associated with Alzheimer's Disease.
Amyloid Plaques: Another hallmark of Alzheimer's Disease.
Neuromelanin-producing Cells (Substantia Nigra): Dopaminergic neurons containing a black pigment.
Corticotropin-releasing hormone (CRH) Neurons (Hypothalamus): Stimulate the pituitary gland to release ACTH.
Thyrotropin-releasing hormone (TRH) Neurons (Hypothalamus): Stimulate the pituitary gland to release TSH.
Olfactory Ensheathing Cells: Support olfactory neuron axons from the nasal cavity to the brain.
Pericytes: Surround endothelial cells in capillaries and venules within the CNS.
Neuroepithelial Stem Cells: Early embryonic neural stem cells.
Cerebrovascular Endothelial Cells: Compose the blood-brain barrier.
Inferior Olivary Neurons: Involved in motor learning and timing.
Parafascicular Nucleus Neurons: Thalamic nucleus involved in pain processing.
Subthalamic Nucleus Neurons: Involved in movement and affected in Parkinson’s disease.
Ependymal Stem Cells: Line the ventricles and can generate neural cells.
Vestibular Hair Cells (Inner Ear): Detect changes in head position.
Clarke's Column Neurons (Spinal Cord): Relay proprioceptive information.
Red Nucleus Neurons: Involved in motor coordination.
Pontine Nuclei Neurons: Relay information between the cortex and cerebellum.
Dentate Nucleus Neurons (Cerebellum): Major output nucleus of the cerebellum.
Cochlear Nucleus Neurons: Process auditory signals from the cochlea.
Tegmental Nucleus Neurons: Located in the brainstem, involved in arousal.
Globus Pallidus Neurons: Part of the basal ganglia circuitry, involved in movement.
Premotor Cortex Neurons: Plan and coordinate motor actions.
Nucleus of the Solitary Tract Neurons: Process visceral sensory information.
Pedunculopontine Nucleus Neurons: Involved in arousal and locomotion.
Cuneate Nucleus Neurons: Relay tactile sensory information from the upper body.
Gracile Nucleus Neurons: Relay tactile sensory information from the lower body.
Central Pattern Generators: Neural circuits producing rhythmic motor patterns.
Accessory Olfactory Bulb Neurons: Process pheromone signals.
Supraoptic Nucleus Neurons: Produce vasopressin.
Nucleus Ambiguus Neurons: Control motor functions of the throat and voice box.
Olivocochlear Neurons: Project from the brain to the cochlea and modulate auditory sensitivity.
Cajal Bodies: Nuclear structures involved in the biogenesis of snRNPs.
Nucleus Raphe Magnus Neurons: Involved in pain modulation.
Parabigeminal Nucleus Neurons: Involved in visual attention.
Ventral Tegmental Area (VTA) Neurons: Produce dopamine, involved in reward.
Pterygopalatine Ganglion Neurons: Involved in salivation and lacrimation.
Lamina I Spinal Neurons: Transmit pain and temperature signals.
On-Off Direction-Selective Ganglion Cells (Retina): Detect directional movement in visual field.
Chromaffin Cells (Adrenal Medulla): Release adrenaline and noradrenaline.
Mauthner Cells (Fish): Mediate escape responses.
Marginal Zone Neurons: Early-born neurons in the cortex.
Barrel Cortex Neurons: Process whisker touch signals in rodents.
Lugaro Cells (Cerebellum): Inhibitory interneurons.
Pulvinar Neurons: Largest thalamic nucleus, involved in visual attention.
Mesencephalic Dopaminergic Neurons: Involved in reward and motor functions.
Cholecystokinin (CCK) Neurons: Release CCK, modulate neural activity.
Incus Cells (Middle Ear): Transmit sound vibrations.
Telencephalic Radial Glial Cells: Generate neurons in the developing telencephalon.
Mossy Fiber Boutons (Cerebellum): Excitatory terminals that form synapses with granule cells.
Ciliated Neural Cells: Motile cilia that help circulate cerebrospinal fluid.
Interstitial Neurons of Cajal: Found in the spinal cord, modulate motor reflexes.
Mesencephalic Trigeminal Neurons: Proprioceptive neurons for the jaw muscles.
Calbindin Neurons: Express calcium-binding protein calbindin, often inhibitory.
Retinal Horizontal Cells: Integrate and regulate input from photoreceptors.
Neurons of the Rostral Ventromedial Medulla: Modulate pain signals.
Medium Afterhyperpolarization (mAHP) Neurons: Characterized by specific post-spike potentials.
Arcuate Fasciculus Neurons: Connect Broca's and Wernicke's areas, crucial for language processing.
Periventricular Nucleus Neurons: Involved in the release of neurohypophysial hormones.
Primary Visual Cortex (V1) Neurons: Process initial visual input from the retina.
Primary Auditory Cortex (A1) Neurons: Process initial auditory input from the cochlea.
Ganglion Cell Layer (Retina): Contains the cell bodies of retinal ganglion cells.
Internal Capsule Neurons: White matter tract connecting the thalamus to the cerebral cortex.
Nucleus of the Trapezoid Body: Involved in auditory processing.
Tectal Neurons: Located in the superior and inferior colliculi; process sensory information.
Neostriatum Neurons: Part of the basal ganglia, involved in movement.
Piriform Cortex Neurons: Involved in olfaction (smell) processing.
Inferotemporal Cortex Neurons: Process complex visual information.
Cingulate Cortex Neurons: Involved in emotion and memory.
Mitral Valve Projection Neurons (Olfactory System): Transmit olfactory information.
Habenular Tract Neurons: Connect habenula to the pineal gland.
Cerebellar Basket Cells: Inhibitory interneurons that regulate Purkinje cells.
Neurolemma Cells: Outermost layer of myelin sheath in peripheral nerves.
Outer Plexiform Layer (Retina): Contains synapses between photoreceptors and bipolar/horizontal cells.
Medial Lemniscus Pathway Neurons: Relay somatosensory information to the thalamus.
Stereocilia: Mechanosensory organelles on hair cells in the inner ear.
Endolymphatic Duct Cells: Part of the inner ear; maintain endolymph balance.
Inferior Colliculus Neurons: Process auditory signals.
Superior Olive Neurons: Process auditory location cues.
Neurosecretory Protein GL Neurons: Modulate neural activity.
Sigmoid Sinus Cells: Part of the vascular system; drain blood from the brain.
Limbic Lobe Neurons: Involved in emotion, behavior, and long-term memory.
Intercalated Cells (Basal Ganglia): Small clusters of neurons between major nuclei.
Centromedian Nucleus Neurons: Part of the thalamus; implicated in Parkinson's and epilepsy.
Prefrontal Cortex Neurons: Involved in complex cognitive behavior and decision-making.
Subplate Neurons: Temporary neurons critical during the early formation of the cerebral cortex.
Fornix Neurons: Major output tract of the hippocampus.
Stria Terminalis: Connects amygdala and hypothalamus.
Corpus Callosum Neurons: Connects the two cerebral hemispheres.
Area Temptata Neurons: Contains thermoreceptors in the hypothalamus.
Nucleus Basalis of Meynert: Forebrain structure involved in acetylcholine production.
Edinger-Westphal Preganglionic Neurons: Control the pupil size and shape of the lens.
Rhomboid Nucleus Neurons: Part of the thalamus; function is still under study.
Spinothalamic Tract Neurons: Transmit pain and temperature sensations.
Anterodorsal Thalamic Nucleus Neurons: Part of the Papez circuit involved in emotion.
Cervical Ganglion Neurons: Part of the sympathetic nervous system.
Dorsal Spinocerebellar Tract Neurons: Convey proprioceptive information to the cerebellum.
Inferior Peduncular Neurons: Connect the cerebellum and the medulla oblongata.
Mamillary Body Neurons: Involved in memory recall.
Ventral Pallidum Neurons: Involved in reward and reinforcement.
Periaqueductal Gray Neurons: Involved in pain modulation and defensive behaviors.
Renshaw Cells: Inhibitory interneurons found in the spinal cord.
Medial Forebrain Bundle Neurons: Connect structures in the basal forebrain and brainstem.
Parahippocampal Gyrus Neurons: Important for encoding and retrieval of memories.
Paramedian Pontine Reticular Formation (PPRF) Neurons: Control horizontal eye movements.
Vomeronasal Organ Neurons: Detect pheromones.
Ansa Lenticularis Neurons: Part of the indirect pathway in the basal ganglia.
Orbitofrontal Cortex Neurons: Involved in decision-making and expectation.
Anterior Cingulate Cortex Neurons: Implicated in emotion and conflict resolution.
Ventral Anterior Nucleus Neurons (Thalamus): Involved in motor planning.
Brodmann Area 25 Neurons: Subgenual cingulate region implicated in mood disorders.
Caudate Nucleus Neurons: Part of the basal ganglia; roles in motor processes and learning.
Putamen Neurons: Involved in a variety of tasks including learning and motor preparation.
Subgenual Anterior Cingulate Cortex Neurons: Implicated in depression.
Hypothalamic Supraoptic Nucleus Neurons: Produce vasopressin.
Postcentral Gyrus Neurons: Main location of the primary somatosensory cortex.
Zona Incerta Neurons: A region of the subthalamus involved in sensory-motor integration.
Globus Pallidus Internus Neurons: Regulate muscular movements.
Mediodorsal Nucleus Neurons (Thalamus): Associated with the prefrontal cortex.
Subcallosal Cortex Neurons: Beneath the corpus callosum, involved in emotion.
Anterior Commissure Neurons: Connects temporal lobes of the two cerebral hemispheres.
Nucleus Accumbens Shell Neurons: Involved in reward and pleasure.
Posterior Parietal Cortex Neurons: Important for sensorimotor integration.
Raphe Magnus Nucleus Neurons: Serotonergic neurons involved in pain modulation.
Midcingulate Cortex Neurons: Roles in pain and cognitive control.
Septohippocampal Nucleus Neurons: Involved in modulating the activity of the hippocampus.
Amygdalohippocampal Area Neurons: Connects the amygdala and hippocampus.
Tectospinal Tract Neurons: Mediates reflex postural movements in response to visual stimuli.
Pedunculopontine Tegmental Nucleus Neurons: Involved in reward, arousal, and movement.
Optic Radiation Neurons: Connects the lateral geniculate nucleus to the visual cortex.
Subthalamus Neurons: Associated with the regulation of movements.
Lingual Gyrus Neurons: Part of the visual cortex.
Retrosplenial Cortex Neurons: Involved in episodic memory and navigation.
Frontal Eye Field Neurons: Control and regulate eye movements.
Dorsomedial Hypothalamic Nucleus Neurons: Involved in the regulation of circadian rhythms.
Hippocampal CA4 Neurons: Part of the hippocampus involved in memory.
Lateral Habenula Neurons: Convey negative reward signals.
Paraventricular Thalamic Nucleus Neurons: Implicated in stress and reward.
Anterior Insula Neurons: Involved in emotion, homeostasis, and self-awareness.
Preoptic Area Neurons: Regulate body temperature and sleep-wake cycles.
Ventral Tegmental Area (VTA) GABAergic Neurons: Inhibit dopaminergic VTA neurons.
Subcommissural Organ Neurons: Secrets proteins into the cerebrospinal fluid.
Pallidum Neurons: Involved in the control of voluntary movement.
Superior Temporal Gyrus Neurons: Involved in auditory processing.
Medial Globus Pallidus Neurons: Regulate thalamic activity.
Arcuate Nucleus Pro-opiomelanocortin (POMC) Neurons: Regulate food intake.
Pre-Bötzinger Complex Neurons: Generate the rhythm of breathing.
Angular Gyrus Neurons: Involved in language, number processing, and spatial cognition.
Medial Septal Neurons: Play a role in the modulation of the hippocampus.
Nucleus Reuniens Neurons: Connects the thalamus and the hippocampus.
Centromedian Parafascicular Complex Neurons: Thalamic nucleus involved in attention and arousal.
Perirhinal Cortex Neurons: Involved in object recognition memory.
Entorhinal Cortex Layer II Stellate Cells: Key in spatial navigation.
Dorsal Cochlear Nucleus Neurons: Process auditory signals.
Mammillothalamic Tract Neurons: Connects the mammillary bodies to the anterior thalamic nuclei.
Intralaminar Nuclei Neurons: Thalamic nuclei involved in arousal and pain perception.
Paracentral Lobule Neurons: Involved in sensorimotor tasks.



Last edited by Otangelo on Wed Oct 04, 2023 8:13 am; edited 2 times in total

https://reasonandscience.catsboard.com

11Evolution of the brain Empty Re: Evolution of the brain Wed Oct 04, 2023 7:52 am

Otangelo


Admin

Arcuate Nucleus Neurons: Key neurons in hypothalamus involved in feeding and hormone release.
Bed Nucleus of the Stria Terminalis Neurons: Involved in stress response and connecting the amygdala to the rest of the brain.
Posterior Cingulate Cortex Neurons: Involved in consciousness and evaluating the internal state.
Dorsal Tenia Tecta Neurons: Play roles in olfactory processing and social memory.
Primary Motor Cortex Neurons (M1): Directly responsible for voluntary movements.
Ventral Posterolateral Nucleus Neurons: Relay somatosensory information to the cerebral cortex.
Supplementary Motor Area Neurons: Involved in the planning of complex coordinated movements.
Olfactory Tubercle Neurons: Play a role in olfactory processing and reward behavior.
Parahippocampal Gyrus Neurons: Important for the encoding and retrieval of memories.
Precentral Gyrus Neurons: The site of the primary motor cortex.
Caudate Head Neurons: Involved in motor processes and procedural learning.
Anterior Insular Cortex Neurons: Associated with emotion, homeostasis, and self-awareness.
Red Nucleus Neurons: Important for motor coordination, especially in the upper limbs.
Substantia Innominata Neurons: Part of the basal forebrain and has diverse roles including involvement in sleep.
Tegmental Nuclei Neurons: A variety of functions from motor to arousal.
Lateral Orbitofrontal Cortex Neurons: Implicated in evaluating outcomes and adaptive learning.
Ventral Tegmental Area Dopaminergic Neurons: Key part of the brain's reward system.
Suprachiasmatic Nucleus Neurons: The brain's main circadian rhythm pacemaker.
Medial Geniculate Nucleus Neurons: Relays auditory information from the inner ear to the primary auditory cortex.
Central Nucleus of the Inferior Colliculus Neurons: Major role in the auditory pathways.
Pineal Gland Pinealocytes: Produce melatonin, which modulates sleep.
Posterior Pituitary Neurohypophysial Cells: Store and release oxytocin and vasopressin.
External Cuneate Nucleus Neurons: Process proprioceptive information from the body.
S1 Barrel Field Neurons: Process tactile information in rodents, especially from whiskers.
Inferior Temporal Cortex Neurons: Involved in visual object recognition.
Locus Coeruleus Noradrenergic Neurons: Main source of noradrenaline in the brain, involved in arousal and alertness.
Claustrum Neurons: A thin sheet of neurons deep within the cortex, function is still being explored.
Fusiform Face Area Neurons: Specialized for facial recognition.
Primary Gustatory Cortex Neurons: Initial cortical processing of taste information.
Secondary Motor Cortex Neurons: Involved in the planning and control of movement.
Dorsal Respiratory Group Neurons in the Medulla: Involved in the basic rhythm of respiration.
Lateral Septal Nucleus Neurons: Involved in reward and reinforcement.
Thalamic Reticular Nucleus Neurons: Modulates activity between the cortex and thalamus.
Central Amygdala Neurons: Involved in processing threats and fear-related behaviors.
Anterior Olfactory Nucleus Neurons: First relay in the olfactory system.
Ventral Posteromedial Nucleus Neurons: Thalamic relay for somatosensory information from the face.
Choroid Plexus Epithelial Cells: Produce cerebrospinal fluid.
Tectal Neurons: Part of the midbrain involved in auditory and visual reflexes.
Ventromedial Prefrontal Cortex Neurons: Decision-making, risk, and reward evaluation.
Anterior Nucleus of Thalamus Neurons: Part of the limbic system, involved in emotion and memory.
Culmen Neurons: A part of the cerebellar vermis involved in motor coordination.
Hippocampal Sulcus Neurons: A groove in the hippocampus, the structure's role in memory.
Ventral Posterior Nucleus Neurons: Major relay nucleus in the thalamus of the somatosensory system.
Spinal Trigeminal Nucleus Neurons: Mediates tactile, pain, and temperature sensation from the face.
Cingulate Sulcus Neurons: A sulcus in the brain associated with the cingulate cortex and its function.
Ventral Tegmental Area GABA Neurons: Inhibitory neurons regulating the brain's reward system.
Basomedial Amygdala Neurons: Involved in modulating memory consolidation.
Cuneus Neurons: Part of the visual processing system.
Nucleus Basalis of Meynert Neurons: Involved in attention and arousal, affected in Alzheimer's disease.
Lateral Periaqueductal Gray Neurons: Role in defensive and fear behaviors, and in pain modulation.
3.Pyramidal Cells: Excitatory neurons involved in higher cognitive functions.
4.Interneurons: Inhibitory neurons that regulate neural activity and network dynamics.
5.Motor Neurons: Connect to muscles and command voluntary movements.
6.Sensory Neurons: Receive sensory input and relay it to the brain.
7.Dopaminergic Neurons: Produce dopamine and play a role in reward and motor control.
8.Serotonergic Neurons: Release serotonin, influencing mood, sleep, and appetite.
9.Cholinergic Neurons: Release acetylcholine, involved in memory and arousal.
10.GABAergic Interneurons: Throughout the Brain, inhibit neural activity and maintain balance.
11.Glutamatergic Neurons: Throughout the Brain, release glutamate, a major excitatory neurotransmitter.
12.Norepinephrine Neurons: Release norepinephrine, affecting alertness and stress response.
13.Astrocytes: Throughout the Brain, support neurons and regulate synaptic transmission.
14.Microglia: Throughout the Brain, immune cells of the brain, involved in defense and repair.
15.Oligodendrocytes: Throughout the Brain, produce myelin, facilitating electrical signal transmission.
16.Basket Cells: Cerebellum and Hippocampus, inhibit neighboring neurons and regulate information flow.
17.Reticular Activating System Neurons: Brainstem, control wakefulness and arousal.
18.Pyramidal Tract Neurons: Cerebral Cortex, send motor signals to the spinal cord and muscles.
19.Granule Cells: Dentate Gyrus, process sensory information in the hippocampus.
20.Pacinian Corpuscle Sensory Neurons: Peripheral Nervous System, detect deep pressure and vibration.
21.NMDA Receptors Neurons: Throughout the Brain, play key roles in synaptic plasticity and memory.
22.Fast-spiking interneurons: Throughout the Brain, known for their rapid action potentials, involved in network timing.
23.Schwann Cells: Peripheral Nervous System, produce myelin sheaths around axons, facilitating signal transmission.
24.Mossy Fibers: Cerebellum and Hippocampus, axons that transmit excitatory information.
25.Climbing Fibers: Cerebellum, originate from the inferior olivary nucleus and synapse onto Purkinje cells.
26.Mirror Neurons: Various Brain Regions, fire both when an individual performs an action and when observing the same action.
27.Medium Spiny Neurons: Striatum, involved in movement and reward-based learning.
28.Horizontal Cells: Retina, regulate the input from photoreceptor cells.
29.Amacrine Cells: Retina, integrate visual information before it's sent to the brain.
30.Ganglion Cells: Retina, their axons form the optic nerve, transmitting visual information to the brain.
31.Photoreceptor Cells: Retina, detect light, including rods (low light) and cones (color).
32.Spindle Neurons: Frontal Cortex, associated with social cognition and intuition.
33.Ependymal Cells: Throughout the Brain, line the ventricles and produce cerebrospinal fluid.
34.Tanycytes: Hypothalamus, involved in neurosecretion and metabolic regulation.
35.Stellate Cells: Cerebral Cortex, star-shaped interneurons that regulate synaptic activity.
36.Lugaro Cells: Cerebellum, interneurons that regulate the activity of Purkinje cells.
37.Golgi Cells: Cerebellum, inhibit granule cells, modulating information flow.
38.Hair Cells: Inner Ear, detect vibrations and are essential for hearing.
39.Merkel Cells: Skin, sensory receptors for touch.
40.Satellite Cells: Peripheral Nervous System, surround and support neuron cell bodies.
41.Von Economo Neurons: Insula and Anterior Cingulate, involved in intuitive reasoning and social emotions.
42.Nociceptors: Peripheral Nervous System, detect pain or potentially damaging stimuli.
43.Thermoreceptors: Peripheral Nervous System, detect changes in temperature.
44.Chemoreceptors: Peripheral Nervous System, detect changes in the chemical composition of the body.
45.Unipolar Brush Cells: Cerebellum, modulate afferent input to the cerebellar cortex.
46.Martinotti Cells: Cerebral Cortex, interneurons that inhibit pyramidal cells.
47.Betz Cells: Motor Cortex, large pyramidal neurons that send motor commands.
48.Chandelier Cells: Cerebral Cortex, inhibitory neurons known for their distinctive axonal structures.
49.Nucleus Accumbens Medium Spiny Neurons: Play a central role in reward and addiction.
50.Vibrissal Sensitive Motor Neurons: Control movements of whiskers in rodents for tactile sensing.

https://reasonandscience.catsboard.com

Sponsored content



Back to top  Message [Page 1 of 1]

Permissions in this forum:
You cannot reply to topics in this forum