SlideShare a Scribd company logo
1 of 136
1-Cytoskeletal components of the neurons and their functions during axon
regeneration.

       Neurons contain a cytoskeleton consisting of neurofibrils, which determine the
shape of the soma and the various processes extending from it, and which transport
substances throught the neuron

 Microtubules and microtubule associated proteins (MAP)

    25-28nm
    are narrow longitudinal tubes present in all neuronal processes. The
       tubesmantain shape, and also transport molecules such as neuro-transmitters
       from the soma to the axon terminals (anterograd transport), or from the terminals
       to the soma (retrograde transport). There are at least two types of axonal
       transport:
   (a) rapid, 400nm
    (b) slow, at less than 1 mm day

Microfilaments

    5-7 nm
    present in high concentrations as a meshwork beneath the membrane of the
     axon. Actin is a important protein in axon development , and causes the
     movement of the growth cone.
    Are a proliferation of globulin actin (G-actin) in the axolemma of the synapse,
     depolarisation of the microfilaments is a prerequesite for releasing of the
     trasmitters from the synapse vesicles.

   Neurofilamets

    10nm
    Are the most abundant of the fibrillar elements in the neuron, and form the
     bones of the cytoskeleton
    they are largely spaced, having tide arms that keep them appart and provide
     neuronal stability
    seem to be invoolved in the transport mechanisms



      3 different types of neurons:
   1. unipolar
   2. bipolar – concentrated mainly in periphery
   3. multi-polar – formed by alfa and beta tubules

     according to size:
 1. Type I Golgi (large+long axons)
 2. Type II Golgi (small+short axons)
2-Types of axoplasmatic transports according direction and speed, their
functions in the intact neuron and during axon regeneration.

Axonal Transport: Various secretory products produced in the cell body are carried to the axon
terminals by special transport mechanism as in the same manner various constituents are carried
from the axon terminals to the cell body.

Three main types of axonal transport are:

⇒   fast anterograde transport

⇒   slow anterograde transport

⇒   fast retrograde transport.


Fast anterograde transport is involved in the transport of materials that have a
functional role at the nerve terminals (e.g., precursors of peptide neurotransmitters,
enzymes needed for the synthesis of small molecule neurotransmitters, and
glycoproteins needed for reconstitution of the plasma membrane) are trasported from
the cell body to the terminals. Polypeptides much larger than final peptide
neurotransmitters (pre-propeptides) and enzymes needed for the synthesis of small
molecule neurotransmitters are synthesized in the rough endoplasmic reticulum. The
vesicles formed in the Golgi apparatus for the axon terminals then become attached to
the microtubules and are transported by fast axonal transport (at a rate of 400 mm/d)
into the nerve terminal.
        The rapid axonal transport depends on the microtubules. The microtubule
provides a stationary track and a microtubule-associated ATPase (kinesin) forms a
cross-bridge between the organelle to be moved and the microtubule. On one end,
kinesin contains two globular heads that bind to the microtubule, and on the other end, it
has a fan-shaped tail that binds to the surface of an organelle. The organelle then moves
by sliding of the kinesin molecule along the microtubule
Slow anterograde transport involves movement of neurofilaments and microtubules
synthesized in the cell body to the terminals at a rate of 5 mm/d. Soluble proteins
transported by this mechanism include actin, tubulin (which polymerizes to form
microtubules), proteins that make up neurofilaments, myosin, and a calcium-binding
protein (calmodulin).
Fast retrograde transport is slower than the fast anterograde transport (about 200 mm/
d). Rapid retrograde transport carries materials from the nerve terminals to the cell
body; the transported materials travel along microtubules.
        An example of transport by this mechanism is nerve growth factor (NGF), a
peptide synthesized by a target cell and transported into certain neurons in order to
stimulate their growth. Materials lying outside the axon terminals are taken up by
endocytosis and transported to the cell body. Fast retrograde axonal transport is also
involved in some pathological conditions. For example, the herpes simplex, polio, and
rabies viruses and tetanus toxin are taken up by the axon terminals in peripheral nerves
and carried to their cell bodies in the central nervous system (CNS) by rapid retrograde
transport.
3-Types of the neuronal synapses, their classification according morphology and
                 neurotransmitters.

                 Synapses needed for communication e.g.interneuronal synapses or intermuscular

                  Direct synapses in neuromuscular junction

                  Indirect synapses in autonomic postganglionic synapse

                   Synapses may be either:

                    1. interneuronal connections

                    2. connections between the neuron and effector



                    Interneuronal connections

according to position of connections:

                      - axodendritic and axo somatic synapses

                      - axo-axonal and dendro – dendritic synapses

according to effect on postsynaptic element

                        -excitatory synapses

                        -inhibitory synapses

according to transmission of the excitation

                        - eletric synapses: - reduced extracellular space

                                            - transport of info by ionic flow

                                            -continuity of cytoplasm

                         -chemical synapses: -presynaptic termination

                                               - postsynaptic element

                                               - active zone

                                               - synaptic cleft – no continuity between pre and post-
                                          synapses

                                               -increased extracellular space
- transport of info by neurotransmitters

4-Glial cell types and their participation in the ontogenetic development of the
CNS


The supporting cells located in the CNS are called neuroglia or simply glial cells. They
are nonexcitable and more numerous (5 to 10 times) than neurons. Neuroglia have been
classified into the following groups: astrocytes, oligodendrocytes, microglia, and
ependymal cells.


       ⇒    Astrocytes


       Among the glial cells, astrocytes are the largest and have a stellate (star-shaped)
appearance because their processes extend in all directions. Their nuclei are ovoid and
centrally located. The astrocytes provide support for the neurons, a barrier against the
spread of transmitters from synapses, and insulation to prevent electrical activity of one
neuron from affecting the activity of a neighboring neuron.
       They are further subdivided into the following subgroups:
a) Protoplasmic Astrocytes: These cells are present in the gray matter in close
association with neurons. Because of their close association with the neurons, they are
considered satellite cells and serve as metabolic intermediaries for neurons. They give
out thicker and shorter processes, which branch profusely. Several of their processes
terminate in expansions called end-feet. Abutting of processes of protoplasmic
astrocytes on the capillaries as perivascular end-feet is one of the anatomical features of
the blood-brain barrier
b) Fibrous Astrocytes
These glial cells are found primarily in the white matter between nerve fibers. Several
thin, long, and smooth processes arise from the cell body; these processes show little
branching. Fibrous astrocytes function to repair damaged tissue, and this process may
result in scar formation.
c) MĂĽller Cells
These modified astrocytes are present in the retina.
⇒   Oligodendrocytes


       These cells are smaller than astrocytes and have fewer and shorter branches.
Their cytoplasm contains the usual organelles (e.g., ribosomes, mitochondria, and
microtubules), but they do not contain neurofilaments. In the white matter,
oligodendrocytes are located in rows along myelinated fibers and are known as
interfascicular oligodendrocytes. These oligodendrocytes are involved in the
myelination process. The oligodendrocytes present in the gray matter are called
perineural oligodendrocytes.


       ⇒   Microglia


       These are the smallest of the glial cells. They usually have a few short branching
processes with thorn-like endings. These processes arising from the cell body give off
numerous spine-like projections. They are scattered throughout the nervous system.
When the CNS is injured, the microglia become enlarged, mobile, and phagocytic.




       ⇒   Ependymal Cells


       Ependymal cells consist of three types of cells:


a) Ependymocytes are cuboidal or columnar cells that form a single layer of lining in
the brain ventricles and the central canal of the spinal cord. They possess microvilli and
cilia. The presence of microvilli indicates that these cells may have some absorptive
function. The movement of their cilia facilitates the flow of the cerebrospinal fluid.
b) Tanycytes are specialized ependymal cells that are found in the floor of the third
ventricle, and their processes extend into the brain tissue where they are juxtaposed to
blood vessels and neurons. Tanycytes have been implicated in the transport of hormones
from the CSF to capillaries of portal system and from hypothalamic neurons to the CSF.
c) Choroidal epithelial cells are modified ependymal cells. They are present in the
choroid plexus and are involved in the production and secretion of CSF. They have tight
junctions that prevent the CSF from spreading to the adjacent tissues

       ⇒   PNS


               In the peripheral nervous system (PNS), Schwann cells provide myelin
       sheaths around axons. The myelin sheaths are interrupted along the length of the
       axons at regular intervals at the nodes of Ranvier. Thus, the nodes of Ranvier are
                                                                                        +
       uninsulated and have a lower resistance. These nodes of Ranvier are rich in Na
       channels, and the action potential becomes regenerated at these regions.
       Therefore, the action potential traveling along the length of the axon jumps from
       one node of Ranvier to another. This type of propagation enables the action
       potential to conduct rapidly and is known as saltatory conduction. During the
       myelination, the axon comes in contact with the Schwann cell, which then
       rotates around the axon in clockwise or counterclockwise fashion. As the
       Schwann cell wraps around the axon, the cytoplasm becomes progressively
       reduced, and the inner layers of the plasma membrane come in contact and fuse
       together.




       ⇒   CNS


       Within the brain and the spinal cord, oligodendrocytes form the myelin sheaths
around axons of neurons. Several glial processes arise from one oligodendrocyte and
wrap around a portion of the axon. The intervals between adjacent oligodendrocytes are
devoid of myelin sheaths and are called the nodes of Ranvier. Unlike in peripheral
axons, the process of an oligodendrocyte does not rotate spirally on the axon. Instead, it
may wrap around the length of the axon. The cytoplasm is reduced progressively, and
the sheath consists of concentric layers of plasma membrane. Unlike in peripheral
nerves, one oligodendrocyte forms myelin sheaths around numerous (as many as 60)
axons of diverse origins.
5-Glial cell types and their involvement in the ontogenetic development of the PNS.

    Glial cells of PNS originate from neural crest cells (plate)

    Schwmann cells produce myelin sheaths around myelinated axons of PNS neurons.

    Glial cells provide support as well as protection for neurons

    Most glial derived from ectodermal tissue (particularly neural tube and crest)

    The exception is microglia- derive from mesoderm .

    sattelite cells surrond neuronal cell bodies in PNS

    Schwmann cells of PNS promote regeneration of peripheral neurons

       A myelinated nerve fiber is one that is surronded by a myelin sheath. In the CNS the
supporting cell is called oligodendrocytes, in the PNS is called Schmann cell.

       The development of the myelin sheath provides an indication of the construction
of its lamellae. The body of Schmann cell forms a rolled up sheet of paper in which the
axon become embebedded. The structure develops, its margins become approximated
and eventually meet together, which results in duplication of the cell membrane – the
mesaxon (pair of parallel plasma membranes of a Schwann cell, marking the point of
edge-to-edge contact by the Schwann cell encircling the axon). This becomes spirally
bound around the axon, probably big movement of the Scwmann cell around the
enclosed axon. The beginning of the duplication lies on the inner side of the myelin
sheath (inner mesaxon) and its end on the outer side (outer mesaxon). Another type of
supporting cell are the satellite cells. Both Schwmann cellas and satellite cells develop
from neural crest cells.

       Myelinating Schwann cells begin to form the myelin sheath in mammals during
fetal development and work by spiraling around the axon, sometimes with as many as
100 revolutions. A well-developed Schwann cell is shaped like a rolled-up sheet of
paper, with layers of myelin in between each coil. The inner layers of the wrapping,
which are predominantly membrane material, form the myelin sheath while the
outermost layer of nucleated cytoplasm forms the neurolemma. Only a small volume of
residual cytoplasm communicates the inner from the outer layers. This is seen
histologically as the Schmidt-Lantermann Incisure. Since each Schwann cell can cover
about a millimeter (0.04 inches) along the axon, hundreds and often thousands are
needed to completely cover an axon, which can sometimes span the length of a body.

6-Describe Wallerian degeneration and different reactions of the glial cells in
CNS and PNS following injury.


       ⇒       Neuronal Injury/ Injury of the Neuronal Cell Body

       The neuronal cell body may be damaged by disease, ischemia (lack of blood
supply), or trauma.
       In the CNS (the brain and spinal cord), the debris produced by neuronal damage
is phagocytosed by microglia. The adjacent fibrous astrocytes proliferate, and the
neurons are replaced by scar tissue.
       In the PNS, macrophages are responsible for the removal of the debris produced
by neuronal damage, and the scar tissue is produced by the proliferation of the
fibroblasts.
       Necrotic cell death is caused by acute traumatic injury that involves rapid lysis
of cell membranes. Necrotic cell death is different from apoptosis. Apoptosis is defined
as a genetically determined process of cell death and is characterized by shrinkage of
the cell, cellular fragmentation, and condensation of the chromatin. During the process
of formation of tissues from undifferentiated germinal cells in the embryo
(histogenesis), more neurons (about 2 times more) are formed than the neurons present
in the mature brain. The excess number of neurons is destroyed during the development
by apoptosis. The mechanism of apoptosis involves activation of a latent biochemical
pathway that is present in neurons and other cells of the body. The cellular debris after
neuronal cell death is removed by phagocytosis, which involves transport of solid
material into the cells (e.g., microglia) that remove the debris by indentation of the cell
membrane of the phagocyte and formation of a vesicle. Pinocytosis is similar to
phagocytosis, except that liquid material is removed. Exocytosis involves fusion of a
vesicle inside the nerve terminal (e.g., a vesicle containing a neurotransmitter) with the
plasma membrane and transportation of the contents of the vesicle outside the nerve
terminal.
⇒   Axonal Damage/ Wallerian Degeneration


       This type of degeneration refers to the changes that occur distally to the site of
damage on an axon. Because protein synthesis occurs primarily in the neuronal cell
body, the segment distal to the damaged site on the axon is affected profoundly.
Initially, the axon swells up and becomes irregular. Later, the axon and the terminal are
broken down into fragments that are phagocytosed by adjacent macrophages and
Schwann cells. Myelin is converted into fine drops of lipid material in the Schwann
cells and is extruded from these cells; it is removed by macrophages in the PNS and
microglial cells and invading macrophages in the CNS. Alterations may also be present
in the proximal segment of the axon up to the first node of Ranvier.


       ⇒   Chromatolysis


       Sectioning of an axon may produce changes in the cell body, and if the injury is
close to the cell body, the neuron may degenerate. The cell body swells up due to edema
and becomes round in appearance, and the Nissl substance gets distributed throughout
the cytoplasm. The nucleus moves from its central position to the periphery due to
edema. The degenerative changes start within hours and are complete within a relatively
short time (about a week).


       ⇒   Anterograde Transneuronal Degeneration
       This type of degeneration occurs in the CNS when damage to a neuron results in
the degeneration of another postsynaptic neuron closely associated with the same
function. For example, damage to an optic nerve results in the degeneration of the
lateral geniculate neurons receiving inputs from this nerve.
       ⇒   Retrograde Transneuronal Degeneration
       This type of degeneration occurs in neurons sending inputs to an injured neuron.
In this situation, terminals of the neuron synapsing with a chromatolytic neuron
withdraw and are replaced by processes of glial cells. The neuron, from which the
inputs to the chromatolytic neuron arise, eventually degenerates.
⇒   Recovery of Neuronal Injury (Regeneration)


       If the damage to the neurons is not severe and they survive the injury,
regeneration is possible, but complete recovery may take as long as 3 to 6 months.
Within about 3 weeks, the swelling of the cell subsides, the nucleus occupies a central
position in the cell body again, and the Nissl bodies are normally distinguished. These
events indicate that protein synthesis has been restored in the neuronal cell body. In
severe damage, although sprouting occurs in axons in the CNS, this process ceases
within a short time (about 2 weeks). In this situation, normal functions of the neurons in
the CNS are not restored. However, in peripheral nerves, an axon can regenerate
satisfactorily if the endoneurial sheaths are intact. In this situation, the regenerating
axons reach the correct destination, and the chances of recovery of function are
reasonable. The growth rate of an axon has been estimated to be 2 to 4 mm per day.
7-Describe developmental zones of the neural tube during histogenesis of the CNS, describe

cell populations originating from the neural crest.


       The nervous system develops from ectoderm, the surface layer of embryonic
tissue. By the third to fourth week of embryonic development, the notochord, of
mesodermal origin, induces the development of the neural plate. By the third to fourth
week of embryonic development, there is a high rate of cell proliferation. As such, the
anterior part of the notochord (of mesodermal origin) begins to thicken, and thus, the
neural plate is formed by the third week of fetal life. The neural plate continues to
thicken over the following week and expands laterally. As it expands, the faster growing
lateral edges of the plate accumulate in a dorsal position as neural folds. As this plate
grows and widens, it forms a shallow groove along its longitudinal axis known as the
neural groove. The posterior end of the neural plate, which is narrower than the anterior
end, will ultimately become the spinal cord, whereas the broader, anterior end will
become the brain. As this plate grows and widens, the neural groove becomes deeper. In
the process of its forming and deepening, some of the cells located in the lateral margin
of the neural groove separate and migrate to a dorsal position to become the neural
crest. As the embryo grows, the neural folds fuse along the midline, thus forming a
neural tube.
       The neural tube consists of three layers:


   1. an inner layer called the ventricular layer, which is in contact with the cavity
       of the neural tube;
   2. an intermediate layer called the mantle layer; and
   3. an outer layer called the marginal layer.


   The ventricular zone is the major proliferative layer and also the first layer of the
forming neural tube to appear. The second layer to form is the marginal layer, followed
by the mantle layer. Early in development, the wall of the neural canal becomes
thickened, in part, by the formation of young or immature neurons that have yet to
completely differentiate (sometimes called neurocytes) in the mantle layer. Because this
layer contains the primary cell bodies of neurons, it will ultimately become the gray
matter of the spinal cord. Axons associated with cells in the mantle layer will grow into
the marginal layer.


       Histogenesis of CNS:

    1. Ventricular zone

       -separates precursors for neurons and glial cells

−       -migration of nuclei to base of ventricular cells

     2. Marginal zone

−       -no cell bodies

        - axons of neurons from intermediate zone invade this zone

−    3. Intermediate zone

−       - forms a interface of ventricular and intermediate zones

         - no migration of nuclei




       Cell populations originating from neural crest:


     During folding of neural crest, groups of cells appear along neural groove,
      neural crest cells
     Some of these cells give rise sensory ganglia (dorsal root ganglia)
     cells of neural crest differentiate into Schwmann cells , pigment cells, meninges
      and odontoblasts.
8-Describe trophic interactions among the neurons and their target tissue, describe
general features of neurotrophic factors.


       Trophism refers to the ability of certain molecules called trophic (nutritional)
factors, to promote cell survival. Neurotrophic factors are polypeptides that support
survival, growth, regeneration, and plasticity of neurons. Most types of neuron are
generated in excessive numbers, followed later by the death of “surplus” cells soon after
axons reach the vicinity of their target. This type of neuronal cell death is regarded to be
a consequence of the competition for the limited amount of neurotrophic factors
released by target cells (e, g., embryonic muscle cells). This is an adaptive means of
adjusting the number of neurons of each type to the number of target cells to be
innervated. The “trophic effect” exerted on neurons is illustrated by the trophic
influences of “taste nerve fibers” upon the taste buds. Not only do the gustatory
nerve fibers convey taste information, but they also have critical roles in both the
maintenance and regeneration of taste buds. Following transection of the gustatory
nerve fibers, the taste buds degenerate. In time, if and when the transected fibers
regenerate into the oral epithelium, new functional taste buds will differentiate from
epithelial cells, Presumably, only taste fibers elaborate the essential trophic factors to
induce the formation of new taste buds from the oral epithelium. Trophic activity could
occur at any time from embryonic life through adulthood. Although a progressive
reduction in activity occurs with age, it is never completely lost. In addition to trophic
effects, there are tropic effects. Tropism refers to the ability of certain molecules to
promote or to guide the outgrowth and directional growth or extension of neuronal
processes (axons and dendrites). Neurotrophins are a class among many neurotrophic
factors that have important roles in the survival of neurons and have widespread effects
throughout the CNS and peripheral nervous system (PNS). Neurotrophins include nerve
growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin 3 (NT3),
and neurotrophin 4/5 (NT 4/5). Examples of related trophic factors include fibroblast
growth factor (FGF), the epidermal growth factor family (EGF) and cytokines. The
cytokines (e.g., interleukin, a leukemia-inhibitory factor) are extracellular or membrane-
anchored polypeptides that mediate communication between cells via cell surface
receptors. Trophic factors, as indicated, have roles in promoting the successive stages in
the cycle of neuronal differentiation, growth, survival, and programmed cell death
       9-Selective neuronal death during ontogenic development of the nervous
system; describe a mechanism and its significance.


       Programme cell death is an integral part of animal tissue development.
Development of an organ or tissue is often preceded by the extensive division and
differentiation of particular cell, with the resultant mass is then arranged in the correct
form with help of apoptosis.

       Unlike cellular death (caused by hypoxia or other various injury), apoptosis
results in cell shrinkage and fragmentation. This allows the cells to be efficiently
phagocytosed and ther components removed without releasing of potentially harmful
intracellular substances into the surrounding tissue.

       If a neuron (and especially its DNA) gets damaged by a virus or a toxin,
apoptosis destroys and silently removes these sick genes, which may serve to protect
surrounding healthy neurons. More importantly, apoptosis is a natural part of
development of the immature central nervous system. One of the many wonders of the
brain is the built-in redundancy of neurons early in development. These neurons
compete vigorously to migrate, innervate target neurons, and drink trophic factors
necessary to fuel this process. Apparently, there is survival of the fittest, because up to
50% of many types of neurons normally die in this time of brain maturation. Apoptosis
is a natural mechanism to eliminate the unwanted neurons without making as big a
molecular mess as doing it via necrosis.

       Cell death via apoptosis is a prominent feature in mammalian neural
development. Recent studies into the basic mechanism of apoptosis have revealed
biochemical pathways that control and execute apoptosis in mammalian cells. Protein
factors in these pathways play important roles during development in regulating the
balance between neuronal life and death. Additionally, mounting evidence indicates
such pathways may also be activated during several neurodegenerative diseases,
resulting in improper loss of neurons.
10-Describe molecular mechanisms for axon navigation to the target tissue during
development and regeneration of the nervous system.




 Molecular mechanisms of axon guidance:

   1. Contact inhibition – adhesion (permissive and non-permissive substrates)

   2. Fasciculation (small, involuntary muscle contractions due to spontaneous
       discharges)

   3. Chemotropism (movement caused by chemical stimulus)




       Cellular and molecular mechanisms for regeneration of nervous system:

Different reaction of the CNS and PNS neurons to injury of neural processes.

PNS neurons: anabolic processes due to increased synthesis of cytoskeleton and
membrane proteins and increased synthesis of RNA.

CNS neurons: reaction with distinct regressive mechanisms(atrophy) and destruction
of neurons. Decreased synthesis if the RNA
11-Describe structural components of the hematoencephalic barrier, functional
significance of HEB.

    Meningeal barriers is formed by arachnoid placed between intradural blood
       vessels and CSF in the subarachnoid space.

    Blood-brain barrier, which is located at the interface between the capillary
       wall and brain tissue. The blood-brain barrier consists of:
       (1) endothelial cells lining the capillary wall with tight junctions between them,
       (2) processes of astrocytes abutting on the capillaries as perivascular end-feet,
       (3) a capillary basement membrane.


              This arrangement of different cells or their processes prevents the
       passage of large molecules from the blood into the extracellular space between
       the neurons and neuroglia and forms the anatomical basis of the blood-brain
       barrier.
              One of the beneficial functions of the blood-brain barrier is to prevent
       entry of blood-borne foreign substances into the brain tissue. However, the
       existence of this blood-brain barrier also presents a problem when the goal is to
       deliver drugs into the CNS. In other organs, tight junctions do not exist between
       the neighboring endothelial cells lining the capillaries.


      The capillaries and the ependymal epithelial cells of the choroid plexus form the
       blood-CSF barrier between the blood and CSF. The presence of this barrier
       results in distinct differences of concentration of some molecules in the blood
       and CSF
12-Describe the CNS liquid compartments and their barriers.


       Fluid (liquid) compartments:

      Interstitial fluid: bathing neurons and glial cells within CNS

      CSF: in subarachnoid space and ventricular system.

      Blood: in the meningeal vessels

      Intracellular fluid: in neurons and glial cells

        CSF: There are four main functions of the CSF. (1) The brain and spinal cord
float in the CSF because the specific gravities of these central nervous system (CNS)
structures are approximately the same. This buoyant effect of the CSF results in
reduction of traction exerted upon the nerves and blood vessels connected with the
CNS. (2) The CSF provides a cushioning effect on the CNS and dampens the effects of
trauma. (3) The CSF also serves as a vehicle for removal of metabolites from the CNS.
(4) Under normal conditions, the CSF provides a stable ionic environment for the CNS.
However, the chemical composition of the CSF may change in certain situations such as
administration of drugs that cross the blood-brain barrier.

       The cerebral barriers:

      Meningeal barriers: is formed by arachnoid placed between intradural blood
       vessels and CSF in the subarachnoid space
13- Describe individual mechanisms of transportation throught the
hematoencephalic barrier.

Transport mechanisms of blood brain barrier (BBB)

⇒ Passive diffusion: lipid soluble substances pass throught the barrier, such as O2,
  Co2, barbiturates, alcohol
⇒ Facilitated diffusion: substances that penetrate membranes by carrier or mediated
  mechanisms. This category of transport carries more molecules and rapidly and
  without consumption of enegy.
⇒ Active transport: consumption of energy, and uses carrier molecules. It proceeds
  against a concentration gradient. E.g amino acids or extracellular potassium. Moves
  molecules from the brain and CSF into blood plasma against a concentration
  gradient.
⇒ Transcytosis: is selective transport by means of the coated vesicles that fuse to
  form trasnsendothelial canal, e.g. Fe bind transferin ehich has Fe receptors

       The combination of the specialized cell membrane of the endothelial cells linked
by intercellular tight junctions is the hallmark of the BBB. This duo effectively excludes
by blocking the passage of many substances across the capillary wall. The permeability
property can be enhanced by the state of phosphorylation of the proteins of the cell–cell
adherens junctions. The cadherin proteins of the adherens junctions also act as a sig-
nalling component between endothelial cells through linkages with the cytoskeletal pro-
tein filaments of the endothelial cells. The presence of so few pinocytotic vesicles with-
in the endothelial cells is indicative that the transcellular movement by vesicles across
the BBB (transendocytosis) is both relatively deficient and slow. However, the selective
passage of substances is related to the presence of high concentrations of carriermedi-
ated transport systems that act as transporters for glucose, essential amino acids, other
required nutrients, and macromolecules. These ensure the passage of essential sub-
stances from the blood to the CNSThe combination of the specialized cell membrane of
the endothelial cells linked by intercellular tight junctions is the hallmark of the BBB.
This duo effectively excludes by blocking the passage of many substances across the ca-
pillary wall. The permeability property can be enhanced by the state of phosphorylation
of the proteins of the cell–cell adherens junctions. The cadherin proteins of the adherens
junctions also act as a signalling component between endothelial cells through linkages
with the cytoskeletal protein filaments of the endothelial cells. The presence of so few
pinocytotic vesicles within the endothelial cells is indicative that the transcellular move-
ment by vesicles across the BBB
14- Describe the CNS structures without total hematoencephalic barrier and their
function.



        There are seven structures in the CNS that lack a blood-brain barrier. Called
circumventricular organs, they are the area postrema, pineal body, subcommissural
organ, subfornical organ, organum vasculosum of lamina terminalis (OVLT),
neurohypophysis (the posterior pituitary gland), and the median eminence.
        They lack tight junctions in their capillaries. Instead, they have fenestrated
capillaries, capillary loops, and large perivascular spaces that permit the passage of
larger circulating molecules into the adjacent brain tissue. It is believed that some
circulating hormones consisting of large molecules reach their target areas in the brain
via the circumventricular organs. For example, the subfornical organ lies in the roof of
the third ventricle. Blood-borne angiotensin II reaches the subfornical organ readily
because of the lack of the blood-brain barrier in this organ and induces thirst for overall
regulation of fluid balance and cardiovascular homeostasis.

    Functional effects
⇒ Acts as homeostatic organs: e.g neurohypophyses releases oxitocin and vasopressin
  (ADH)
⇒ Acts as as chemoreceptors e.g. Angiotensin II may act to increase blood pressure
⇒ Acts as osmoreceptors

⇒ The area postrema is the vomiting centre of the brain, detect noxious substances in
  the blood and stimulate vomiting in order to rid that substances from the body
15- Measuring of the cerebral blood flow

        The metabolic demands of the brain must be met with the blood supply to this
organ. Normal cerebral blood flow is about 50 mL/100 g of brain tissue/min. Thus, a
brain of average weight (1500 g) has a normal blood flow of 750 mL/min. Even a brief
interruption of the blood supply to the CNS may result in serious neurological
disturbances. A blood flow of 25 mL/100 g of brain tissue/min constitutes ischemic
penumbra (a dangerously deficient blood supply leading to loss of brain cells). A blood
flow of 8 mL/100 g of brain tissue/min leads to an almost complete loss of functional
neurons. Consciousness is lost within 10 seconds of the cessation of blood supply to the
brain.
        Freks principle:
        Cerebral blood flow can be measured by determining the amount of nitrous
oxide removed from the blood stream (Qx) per unit of time and dividing that value by
the difference between the concentration in the atrial blood (Ax) and the in the venous
blood (Vx):
                                Qx
                       CBF= ---------------
                             [Ax] - [Vx]

Qx= amount of nitrous oxide removed from the blood
Ax= concentration in atrial blood
Vx= concentration in the venous blood


Average blood flow in young adults is 54ml/100g/min
Average brain weight 1400g hence we have a blood flow to brain corresponding to
756ml/min

Factors that affect cerebral blood flow :

   1)   Intracranial pressure
   2)   Blood viscosity
   3)   Mean venous pressure
   4)   Mean atrial pressure
   5)   Constriction and dilation of cerebral arterioles
16 – Blood flow in various parts of the brain Arterial supply of the brain:

        Blood supply to the brain is derived from two arteries: (1) the internal carotid
artery and (2) the vertebral artery. These arteries and their branches arise in pairs that
supply blood to both sides of the brain. The basilar artery is a single artery located in the
midline on the ventral side of the brain. The branches of the basilar artery also arise in
pairs.

       Internal Carotid Artery

       This artery arises from the common carotid artery on each side at the level of the
thyroid cartilage and enters the cranial cavity through the carotid canal.
       Branches:

⇒ The Ophthalmic Artery: enters the orbit through the optic foramen and gives rise
  to the central artery of the retina, which supplies the retina and cranial dura.
  Interruption of blood flow in the ophthalmic artery causes loss of vision in the
  ipsilateral eye.
⇒ The Posterior Communicating Artery: arises at the level of the optic chiasm and
  travels posteriorly to join the posterior cerebral arteries. Small branches arising
  from this artery supply blood to the hypophysis, infundibulum, parts of the
  hypothalamus, thalamus, and hippocampus.
⇒ The Anterior Choroidal Artery: arises near the optic chiasm and supplies the
  choroid plexus located in the inferior horn of the lateral ventricle, the optic tract,
  parts of the internal capsule, hippocampal formation, globus pallidus, and lateral
  portions of the thalamus.
⇒ The Anterior Cerebral Artery: The anterior cerebral artery travels rostrally
  through the interhemispheric fissure. It supplies blood to the medial aspect of the
  cerebral hemisphere, including parts of the frontal and parietal lobes. This artery
  also supplies blood to the postcentral gyrus and precentral gyrus. Occlusion of one
  of the anterior cerebral arteries results in loss of motor control (paralysis) and loss
  of sensation in the contralateral leg. Other structures supplied by the anterior
  cerebral artery include the olfactory bulb and tract, anterior hypothalamus, parts of
  caudate nucleus, internal capsule, putamen, and septal nuclei.

⇒ The Anterior Communicating Artery: at the level of the optic chiasm, the
  anterior communicating artery connects the anterior cerebral arteries on the two
  sides. A group of small arteries arising from the anterior communicating and
  anterior cerebral arteries penetrates the brain tissue almost perpendicularly and
  supplies blood to the anterior hypothalamus, including preoptic and suprachiasmatic
  areas.

⇒ The Medial Striate Artery (Recurrent Artery of Heubner): arises from the
  anterior cerebral artery at the level of the optic chiasm and supplies blood to the
  anteromedial part of the head of the caudate nucleus and parts of the internal
  capsule, putamen, and septal nuclei. The medial striate and the lenticulostriate
  arteries penetrate the perforated substance.
⇒ The Middle Cerebral Artery: at the level just lateral to the optic chiasm. Branches
  of the middle cerebral artery supply blood to the lateral convexity of the cerebral
  hemisphere including parts of the temporal, frontal, parietal, and occipital lobes.
Vertebro-Basilar Circulation:

This system includes the two vertebral arteries, the basilar artery (which is formed by
the union of the two vertebral arteries), and their branches. This arterial system supplies
the medulla, pons, mesencephalon, and cerebellum. Braches:

⇒ The vertebral artery: on each side is the first branch arising from the subclavian
  artery. It enters the transverse foramen of the sixth cervical vertebrae, ascends
  through these foramina in higher vertebra, and eventually enters the cranium
  through the foramen magnum. In the cranium, at the medullary level, each vertebral
  artery gives off the anterior spinal artery, the posterior inferior cerebellar artery, and
  the posterior spinal artery.
⇒ The Anterior Spinal Artery: at the confluence of the two vertebral arteries, two
  small branches arise and join to form a single anterior spinal artery. This artery
  supplies the medial structures of the medulla, which include the pyramids, medial
  lemniscus, medial longitudinal fasciculus, hypoglossal nucleus, and the inferior
  olivary nucleus.
⇒ The Posterior Inferior Cerebellar Artery (PICA): arises from the vertebral artery
  and supplies the regions of the lateral medulla that include the spinothalamic tract,
  dorsal and ventral spinocerebellar tracts, descending sympathetic tract, descending
  tract of cranial nerve V, and nucleus ambiguus.

⇒ The Posterior Spinal Artery (PSA): It is the first branch of the vertebral artery in
  the cranium in about 25% of cases. However, in a majority of cases (75%), it arises
  from the posterior inferior cerebellar artery. In the caudal medulla, this artery
  supplies the fasciculus gracilis and cuneatus as well as the gracile and cuneate
  nuclei, spinal trigeminal nucleus, dorsal and caudal portions of the inferior
  cerebellar peduncle, and portions of the solitary tract and dorsal motor nucleus of
  the vagus nerve.
⇒ The Basilar Artery: The two vertebral arteries join at the caudal border of the pons
  to form the single basilar artery.
⇒ The Anterior Inferior Cerebellar Artery (AICA): is the most caudal branch
  arising from the basilar artery. The AICA supplies the ventral and inferior surface
  of the cerebellum and lateral parts of the pons.
⇒ The labyrinthine (internal auditory) artery: is usually a branch of the AICA and
  supplies the cochlea and labyrinth.
⇒ The Pontine Arteries: Several pairs of pontine arteries arise from the basilar
  artery. Some pontine arteries (the paramedian arteries) enter the pons immediately
  and supply the medial portion of the lower and upper pons. Some pontine arteries
  (the short circumferential arteries) travel a short distance around the pons and
  supply substantia nigra and lateral portions of the midbrain tegmentum.
⇒ The superior cerebellar artery: arises just caudal to the bifurcation of the basilar
  artery and supplies the rostral level of the pons, caudal part of the midbrain, and
  superior surface of the cerebellum.
       ⇒ The posterior cerebral arteries arise at the terminal bifurcation of the
           basilar artery. Branches of the posterior cerebral arteries supply most of the
           midbrain, thalamus, and subthalamic nucleus.
  Cerebral Arterial Circle (Circle of Willis)
The cerebral arterial circle surrounds the optic chiasm and the infundibulum of the
pituitary. It is formed by the anastomosis of the branches of the internal carotid artery
and the terminal branches of the basilar artery. The anterior communicating artery
connects the two anterior cerebral arteries, thus forming a semicircle. The circle is
completed as the posterior communicating arteries arising from the internal carotid
arteries at the level of the optic chiasm travel posteriorly to join the posterior cerebral
arteries that are formed by the bifurcation of the basilar artery. The circle of Willis is
patent in only 20% of individuals. When it is patent, this arterial system supplies the
hypothalamus, hypophysis, infundibulum, thalamus, caudate nucleus, putamen, internal
capsule, globus pallidus, choroid plexus (lateral ventricles), and temporal lobe.
17- Regulation of cerebral circulation. Brain metabolism.


Brain metabolism:

3 metabolic factors have potent effect on control of cerebral blood flow (CBF) :

⇒ Increase in [CO2] leads to increase of CBF.
  CO2 + H2O  H2CO3  HCO3- + H+ AND the H+ causes the dilation of
  cerebral vessels

⇒ Increase in H+ leads to increase in CBF

⇒ Decrease in O2 leads increase CBF via vasodilation


The brain (2% of the total body weight) receives about 15% of the cardiac output and
consumes about 20% of the total O2 consumption.
The brain is highly sensitive to disturbances of the blood supply. Ischaemia lasting
seconds causes symptoms and lasting for few minutes causes irreversible damage.

The caliber of the arterioles is regulated by:

⇒ Local vasodilators; metabolites (e.g. CO2)~
⇒ Vasoactive substances produced by the endothelium, circulating peptides such as
  angiotensin II
⇒ Vasomotor nerves
⇒ Autoregulation mechanisms (the systemic blood pressure increases, but the cerebral
  blood flow remains constant by:
        a) intraluminar pressure within the arterioles elicits direct myogenic
            responde
        b) hypocapnia causes arterial vasoconstriction

Intracranial pressure:

⇒ cerebral vessels are compressed even if there is no increase in intracranial pressure
⇒ any change in venous pressure causes a similar change in intracranial pressure. A
  rise in venous pressure decreases cerebral blood flow.


⇒ Cerebreal circulation has strong sympathetic innervation extending from the
    superior cervical ganglion
⇒ Brain extremely sensitive to hypoxia
⇒ Ammonia very toxic to nerve cells and leaves the brain in the form of glutamine
18- Formation and absorption of cerebrospinal fluid. Function of cerebrospinal
fluid.


       Formation of the Cerebrospinal fluid:

        About 70% of the CSF present in the brain and spinal cord is produced by the
choroid plexuses. The remaining 30% of CSF, which is secreted by the parenchyma of
the brain, crosses the ependyma (a single layer of ciliated columnar epithelial cells
lining the ventricular system) and enters the ventricles. The formation of CSF is an
active process involving the enzyme carbonic anhydrase and specific transport
mechanisms.
        The formation of the CSF first involves filtration of the blood through the
fenestrations of the endothelial cells that line the choroidal capillaries. However, the
movement of peptides, proteins, and other larger molecules from this filtrate into the
CSF is prevented by the tight junctions that exist in the neighboring epithelial cells that
form the outer layer of the choroid plexus. Energy-dependent active transport
mechanisms are present in the choroidal epithelium for transporting Na+ and Mg2+ ions
into the CSF and for removing K+ and Ca2+ ions from the CSF. Water flows across the
epithelium for maintaining the osmotic balance. Normally, the rate of formation of CSF
is about 500 mL/day and the total volume of CSF is 90 to 140 mL, of which about 23
mL is in the ventricles, and the remaining is in the subarachnoid space.

       Circulation:

        The movement of CSF is pulsatile. It flows from the lateral ventricles into the
third ventricle through the foramina of Monro where it mixes with more CSF. Then, it
flows through the cerebral aqueduct (aqueduct of Sylvius) into the fourth ventricle,
where additional CSF is secreted. The fluid leaves the ventricular system via the
foramina of Luschka and Magendie and enters the cerebellomedullary cistern (cisterna
magna). The CSF then travels rostrally over the cerebral hemisphere where it enters the
arachnoid villi.
        Absorption: is made throught the arachidonic vili. The CSF drains into dural
venous sinuses, there are valves here, so fluid flows only from vili to veins where
pressure difference is appropriate.
        Functions:
        There are four main functions of the CSF.
        (1) The brain and spinal cord float in the CSF because the specific gravities of
these central nervous system (CNS) structures are approximately the same. This
buoyant effect of the CSF results in reduction of traction exerted upon the nerves and
blood vessels connected with the CNS.
        (2) The CSF provides a cushioning effect on the CNS and dampens the effects of
trauma. (3) The CSF also serves as a vehicle for removal of metabolites from the CNS.
        (4) Under normal conditions, the CSF provides a stable ionic environment for
the CNS.
        Composition:

Normally, very little protein is present in the CSF, and this is the primary difference
between CSF and blood serum. The concentrations of glucose, as well as
19 – Resting potential of the neuron

        Resting membrane potential: When a neuron is not generating action
potentials, it is at rest. When the neuron is at rest, its cytosol along the inner surface of
its membrane is negatively charged compared with the charge on the outside. Typically,
the resting membrane potential (or resting potential) of a neuron is -65 millivolts (mV).
         The potential difference across the cell membrane during resting state is called
the resting membrane potential. The lipid bilayer of the neuronal membrane maintains
this separation of charges by acting as a barrier to the diffusion of ions across the
membrane. The ion concentration gradients across the neuronal membrane are
established by ion pumps that actively move ions into or out of neurons against their
concentration gradients. The selective permeability of membranes is due to the presence
of ion channels that allow some ions to cross the membrane in the direction of their
concentration gradients. The ion pumps and ion channels work against each other in this
manner. If the neuronal membrane is selectively permeable to only a K+ ion, this ion
will move out of the neuron down its concentration gradient. Therefore, more positive
charges accumulate outside the neuron. The fixed negative charges inside the neuron
impede the efflux of positively charged K+ ions, and excess positive charges outside the
neuron tend to promote influx of the K+ ions into the neuron due to the electrostatic
forces. The opposite charges attract, while similar charges repel each other. Thus, two
forces are acting on the flow of K+ ions out of the neuron; a higher concentration inside
the neuron (concentration gradient) tends to expel them out of the neuron, while the
electrostatic forces tend to prevent their flow out of the neuron.
        When the two opposing forces are equal, K+ concentrations inside and outside
the neuron are in equilibrium. The value of the membrane potential at this time is called
the K+ equilibrium potential. Thus, if the neuronal membrane contained only K+
channels, the resting membrane potential would be determined by the K+ concentration
gradient and would be equal to the equilibrium potential for K+ ions (approximately -80
mV). However, as stated earlier, the resting membrane potential of a neuron is usually
-65 mV. This is because neurons at rest are permeable to the Na+ ion also. The Na+ ions
tend to flow into the neuron due to two forces: (1) concentration gradient of Na+ ions
(extracellular Na+ concentration is much higher than its intracellular concentration) and
(2) electrostatic forces (there is an excess of positive charges outside and an excess of
negative charges inside the neuron). Due to the influx of Na+ ions, the resting membrane
potential deviates from that of the K+ equilibrium potential (i.e., it becomes -65 mV
instead of -80 mV).
        However, the membrane potential does not reach the equilibrium potential for
    +
Na . The reason for the neuron's inability to attain a resting membrane potential closer
to the Na+ equilibrium potential is that the number of open nongated Na+ channels is
much smaller than the number of open nongated K+ channels in the resting state of a
neuron. The permeability of Na+ is small despite large electrostatic and concentration
gradient forces tending to drive it into the neuron. To maintain a steady resting
membrane potential, the separation of charges across the neuronal membrane must be
maintained at a constant. This is accomplished by the Na+-K+ pump described earlier.
        Goldman equation: since the neuronal membrane is permeable to more than
one ion, the goldman equation is used to calculate membrane potential. This equation
takes into account the contribution of the permeability of each ion and its extra- and
intracellular concentration.
        Nerst equation: is used to calculate equilibrium potential of an ion that is
present on both sides of the cell membrane.
20- Receptor, synaptic and action potential-description

Receptor potential: whatever the stimulus that excites the receptor, its immediate
effect is to arrange the membrane potential of the receptor. This change is called
receptor potential.
Different receptors can be excited, either:
           a) By mechanical deformation that stretches the receptor membrane and
               opens ion channels.
           b) By application of a chemical to the membrane
           c) By change of temperature
           d) By eletromagnetic radiation
When the receptor potential rises above the threshold for eliciting an action potential,
the the action potential begins to appear. The more the receptor potential rises above the
threshold level, the greater becomes the action potential frequency

Synaptic potential: an interaction of a transmitter on postsynaptic neuron initiates a
synaptic potential.
Can be either: EPSP (excitatory postsynaptic potential)
                 IPSP (inhibitory postsynaptic potential)
Action potential: rapid change in the membrane potential. It begins with a sudden
change from the normal resting membrane potential to a positive membrane potential.
        When a neuron receives an excitatory input, the neuronal membrane is
depolarized, resulting in an opening of some voltage-gated Na+ channels and influx of
Na+ . The accumulation of positive charges due to influx of Na+ promotes depolarization
of the neuronal membrane. When the membrane potential reaches threshold potential,
the chances of generating an action potential are about 50%. However, when the
membrane is depolarized beyond the threshold potential, a sufficient number of voltage-
gated Na+ channels open, relative permeability of Na+ ions is greater than that of K+
ions, and action potentials are generated with certainty.
        During the rising phase of the action potential, there is a rapid depolarization of
the membrane due to increased permeability of Na+. The depolarization continues so
that the membrane potential approaches the Na+ equilibrium potential. The part of the
action potential where the inside of the neuron is positive relative to the outside is called
the overshoot. Towards the end of the rising phase of the action potential, voltage-gated
Na+ channels are inactivated, and the influx of Na+ through these channels is stopped.
During the falling phase of the action potential, the neuron is repolarized by opening of
voltage-gated K+ channels, which allows increased efflux of K+ from the neuron through
these channels. The opening of voltage-gated K+ channels is also caused by
depolarization of the neuronal membrane. Because these voltage-gated K+ channels
open with a delay (about 1 msec) after the membrane depolarization and their opening
rectifies the membrane potential, they are called delayed rectifier K+ channels. At the
end of the falling phase, the membrane potential is more negative than the resting
potential because of increased K+ permeability caused by the opening of the delayed
rectifier K+ channels in addition to the already present resting K+ permeability through
nongated channels. The permeability is closer to the equilibrium potential of K+ because
there is little Na+ permeability during this period. This portion of the action potential is
called after-hyperpolarization or undershoot. Once after-hyperpolarization has occurred,
the resting membrane potential is restored gradually as the voltage-gated K+ channels
close again.
21 – Ionic basis of membrane potential changes

        The resting membrane potential of a neuron is usually -65 mV. At rest, Na+
influx into the neuron through open nongated Na+ channels is balanced by the efflux of
K+ through open nongated K+ channels. Thus, the membrane potential remains constant
closer (but not equal) to the K+ equilibrium.
        When a neuron receives an excitatory input, the neuronal membrane is
depolarized, resulting in an opening of some voltage-gated Na+ channels and influx of
Na+. Na+ channels are normally closed. The accumulation of positive charges due to
influx of Na+ promotes depolarization of the neuronal membrane. When the membrane
potential reaches threshold potential, the chances of generating an action potential are
about 50%. However, when the membrane is depolarized beyond the threshold
potential, a sufficient number of voltage-gated Na+ channels open, relative permeability
of Na+ ions is greater than that of K+ ions, and action potentials are generated with
certainty. Generation of an action potential is an all-or-nothing phenomenon. Because
the concentration of Na+ channels is relatively high at the axon hillock, this is the site of
generation of action potentials in a neuron.
          During the rising phase of the action potential generation, there is a rapid
depolarization of the membrane due to increased permeability of Na+. The
depolarization continues so that the membrane potential approaches the Na+ equilibrium
potential. The part of the action potential where the inside of the neuron is positive
relative to the outside is called the overshoot. Towards the end of the rising phase of the
action potential, voltage-gated Na+ channels are inactivated, and the influx of Na+
through these channels is stopped.
During the falling phase of the action potential, the neuron is repolarized by opening of
voltage-gated K+ channels, which allows increased efflux of K+ from the neuron through
these channels. The opening of voltage-gated K+ channels is also caused by
depolarization of the neuronal membrane. Because these voltage-gated K+ channels
open with a delay (about 1 msec) after the membrane depolarization and their opening
rectifies the membrane potential, they are called delayed rectifier K+ channels. At the
end of the falling phase, the membrane potential is more negative than the resting
potential because of increased K+ permeability caused by the opening of the delayed
rectifier K+ channels in addition to the already present resting K+ permeability through
nongated channels. The permeability is closer to the equilibrium potential of K+ because
there is little Na+ permeability during this period. This portion of the action potential is
called after-hyperpolarization or undershoot. Once after-hyperpolarization has occurred,
the resting membrane potential is restored gradually as the voltage-gated K+ channels
close again.

   The sodium channel exists in the following three states: resting, activated, or
inactivated.

   •   Resting state: During this state, the activation gate closes the channel pore
       while the inactivation gate is open. With the channel pore closed, Na+ cannot
       flow into the neuron.
   •   Activated state: During the rising phase of action potential, both activation and
       inactivation gates are open, and Na+ ions flow into the neuron.
   •   Inactivated state: During this state, the inactivation gate closes the channel
       pore while the activation gate is still open. Even though the activation gate is
       open, Na+ cannot flow into the neuron. The neuron cannot be activated until the
22- Ion channels in neurons – their distributions

Ion Channels:

.      Ion channels are made up of proteins that are embedded in the lipid bilayer of
the neuronal membrane across which they span. They are characterized by the following
general properties.

   •   The flow of ions through the channels does not require metabolic energy; the
       flow is passive.
   •   The electrochemical driving force across the membrane, but not the channel
       itself, determines the direction and eventual equilibrium of this flow.
   •   The ionic charge determines whether a channel allows an ion to flow through;
       some channels allow cations while others allow anions to flow through them.
   •   Most cation-selective channels allow only one ion species (e.g., Na+ or K+ or
       Ca2+) to flow through them. However, some channels allow more than one ion
       species to flow through them. For example, when L-glutamate (an excitatory
       amino acid neurotransmitter) activates an N-methyl-D-aspartic acid (NMDA)
       receptor, both Na+ and Ca2+ ions flow through the NMDA receptor channel into
       the neuron.
   •   Most anion-selective channels allow only Cl- to flow through them.
   •   Some blockers can prevent the flow of ions through the ion channels. For
       example, phencyclidine (PCP, or Angel Dust) blocks the NMDA receptor
       channel.

    Classification of Ion Channels:
    Nongated Channels:
    Although nongated channels are capable of opening as well as closing, most of the
time they are in the open site. They control the flow of ions during the resting
membrane potential. Examples include nongated Na+ and K+ channels that contribute to
the resting membrane potential.
    Gated Channels:
    These channels are also capable of opening as well as closing. All gated channels
are allosteric proteins
    The channels that are opened or closed by a change in the membrane potential are
called voltage-gated channels. The opening and closing of the channel is believed to be
due to the movement of the charged region of the channel back and forth through the
electrical field of the membrane.
    Voltage-gated channels exist in three states:
    (1) resting state (the channel is closed but can be activated)
    (2) active state (the channel is open), and
    (3) refractory state (the channel is inactivated).
    Changes in the electrical potential difference across the membrane provide the
energy for gating in these channels. Genes encoding for voltage-gated Na+, K+, and Ca2+
channels belong to one family. These channels are described as follows.
    The voltage-gated Na+ channel is formed by a single long polypeptide (a string of
amino acids containing peptide bonds) that has four domains (I-IV). Each domain has
six hydrophobic alpha helices (S1aS6) that span back and forth within the cell
membrane. The four domains join together and form an aqueous pore of the channel.
An additional hydrophobic region connects the S5 and S6 alpha helical segments,
forming a pore loop. The presence of this pore loop makes the channel more permeable
to Na+ than to K+. The membrane-spanning S4 alpha helical segment is believed to be
voltage sensitive. At the resting membrane potential, the channel pore is closed. The S4
segment undergoes a conformational change when the membrane potential changes
(e.g., when the neuron is depolarized), the S4 segment is pushed away from the inner
side of the membrane, and the channel gate opens, allowing an influx of Na + ions. There
are some cases where Na+ permeability is blocked. Tetrodotoxin (TTX), a toxin isolated
from the ovaries of Japanese puffer fish, binds to the sodium channel on the outside and
blocks the sodium permeability pore. Consequently, neurons are not able to generate
action potentials after the application of TTX. These channels are also blocked by local
anesthetic drugs (e.g., lidocaine).
    The basic structure of the voltage-gated Ca2+ channel is similar to that of the
voltage-gated Na+ channel. Ca2+ ions enter the postsynaptic neurons through these
channels and activate enzymes. Depolarization of presynaptic nerve terminals results in
entry of Ca2+ ions into the terminal via these channels. An increase in the levels of
intracellular Ca2+ results in the release of transmitters from presynaptic nerve terminals.
Different varieties of voltage-gated K+ channels have been identified, and they serve
different functions. The general scheme describing the components of this channel is
similar to that of the voltage-gated Na+ channel, except that the voltage-gated K+
channel consists of four polypeptides. It should be recalled that each polypeptide
contributing to the formation of a large protein molecule is called a subunit. Each
subunit of a voltage-gated K+ channel consists of six alpha-helical membrane-spanning
segments (S1 a S6). A pore loop makes the channel more permeable to K + than to Na+.
The S4 segment acts as an activation gate. The K+ channels are generally blocked by
chemicals such as tetraethylammonium (TEA) or 4-aminopyridine.
        The ligand-gated channels are opened by noncovalent binding of chemical
substances with their receptors on the neuronal membrane. These chemical substances
include:
        (1) transmitters or hormones present in the extracellular fluid that bind to their
receptors on the extracellular side of the channel and bring about a conformational
change to open the channel (e.g., acetylcholine, γ-aminobutyric acid [GABA], or
glycine); and
        (2) an intracellular second messenger (e.g., cyclic adenosine monophosphate,
which is activated by a transmitter such as norepinephrine). The second messenger can
open the channel (1) directly by binding to the channel and causing a conformational
change or (2) indirectly by phosphorylating the channel protein in the presence of a
protein kinase and causing a conformational change; this effect on the channel is
reversed by dephosphorylation catalyzed by a protein phosphatase. Genes encoding for
transmitter-gated channels (e.g., channels activated by acetylcholine, GABA, or
glycine) and genes encoding for voltage-gated channels belong to different families.
Mechanically gated channels open by a mechanical stimulus and include the channels
involved in producing generator potentials of stretch and touch receptors.

23 – Spreading of membrane potentials. Length and time constant of the
membrane.

       An action potential elicited at any one point on an excitable membrane usually
excites adjacent portions of the membrane, resulting in the propagation of the action
potential.
A nerve fiber excited at its midportion-develop increased permeability to Na+.
Positive electrical charges carried by the inward diffusing Na+ flow inside the fiber
throught depolarized membrane and then for several milimiters along the core of the
axon. These positive cgarges increase the voltage to above the thereshold. Thus the
depolarization process travels along the entire extent of the fiber and the transmission of
this depolarization process is called nerve impulse. The action potential will travel in
both directions away from the stimulus until the entire membrane becomes depolarized.
        Once an action potential has been elicited at any point on the membtrane of a
normal fiber, the depolarization process travels over the entire membrane if conditions
are right, or it does not travel at all if conditions are not right- this is called “all or
nothing principle”, and it applies to all normal excitable tissues. Occasionally, the action
potential reaches a point on the membrane at which it does not generate voltage to
stimulate the next area of the membrane. When this occurs, the spread of depolarization
stops. Therefore, for continued propagation of an impulse , the ratio of action potential
to threshold for excitation must at all times be greater than 1 – safety factor for
propagation.

       Length-constant: measures the effectiveness of neuron in longitudinal signal
transduction.
                        Rm
               λ= √ -------------------
                       (Ri + Ro )


       SOFIA: lambda e igual a raiz quadrada de abrir parentesis Rm sobre abrir
parentesis ( Ri + Ro )

       Rm: is the resistance across the membrane
       Ri: resistance inside the membrane
       Ro: is the resistance outside the membrane

       The larger the length constant the bigger the effect of the action potential
       Long leght constant results in spatial summation

       Time constant:

            T= Rm . Cm

               R: resistance
               C: capacitance

       Long-time constant results in temporal summation


       24 – Temporal summation of membrane potentials.


       Temporal summation: is an effect generated by a single neuron as a way of
achieving action potential. Summation occurs when the time constant is sufficiently
long and the frequency of rises in potential are high enough that a rise in potential
begins before a previous one ends. The amplitude of the previous potential at the point
where the second begins will algebraically summate, generating a potential that is
overall lager than the individual potentials. This allows the potential to reach the
thereshold to generate the action potential.
        Thus successive postsynaptic potentials caused by discharges from a single pre-
synaptic, if they occur rapidly enough, can summate in the same way that postsynaptic
potentials may summate from widely distributed terminals over the surface of a neuron.
        The degree of temporal summation is directly proportional to time – constant;
and it occurs when the second potential arrises before the 1st has decreased.
        Temporal summation is involved in vision. The inverse proportion of intensity
and time, applies as long as the stimulus is no greater than 0.1 second. For example, at
0.1 second, 130 quanta are absorbed, un any matter of provision, but when raised to 1
second there is a lesser rate of summation, needing 230 quanta to compensate for the
decrease in intensity. The frequency of vision is function of frequency of flashes, so the
longer the stimulus, the better chance it can attain the number of quanta needed for
vision.




       25 – Conduction velocity of the action potential, its determiants.


       When a region of an unmyelinated axonal membrane is depolarized sufficiently
by a depolarizing stimulus (e.g., a synaptic potential in a neuron) to reach a threshold
potential, voltage-gated Na+ channels open, Na+ flows into the axoplasm, and an action
potential is generated in that region of the axon. Some of the current generated by the
action potential spreads by electrotonic conduction (passive spread) to an adjacent
region of the axon. The passive spread of current occurs by movement of electrons, and
movement of Na+ ions is not required. At the adjacent region, the passive spread of
current results in opening of voltage-gated Na+ channels, influx of Na+ into the
axoplasm, and generation of an action potential. In other words, the passive spread of
voltage along the length of an axon results in an active regeneration process.
        The propagation of an action potential along the axon depends on the cable
properties of the axon. The larger the diameter of the axon, the lower the resistance
there is to the flow of current along its length. Therefore, the conduction velocity
(propagation of action potential) along the length of the axon can be increased by
increasing its diameter. For example, the axons of stellate ganglion neurons in the squid
are about 1 mm in diameter (1000 times larger than the axons of mammalian neurons).
The conduction of action potential in these squid giant axons is faster than in
mammalian axons. The squid needs these fast conducting axons for faster contraction of
the mantle muscles that produce a jet propulsion effect needed for quick escape from
predators.
        In vertebrates, the conduction velocity is increased by myelination of axon. A
myelin sheath consists of about 1-mm lengths of as many as 300 concentric layers of
membrane around a single axon. In the peripheral nervous system, myelin is formed by
Schwann cells. In the central nervous system, oligodendrocytes form the myelin. Nodes
of Ranvier (bare segments of the axonal membrane with a very high density of voltage-
gated Na+ channels) are present in between the segments of the myelin sheath. The
myelinated segments of an axon are not excitable and have a high resistance to the
leakage of current across them. On the other hand, passive spread of current can
generate an intense current at the nodes of Ranvier due to the presence of a high density
of voltage-gated Na+ channels.
When a depolarizing stimulus (e.g., a synaptic potential in a neuron) arrives at a node of
Ranvier, Na+ channels open, there is an influx of Na+ ions, and an action potential is
generated at that node. Some current generated by the action potential spreads passively
to the next node of Ranvier, and depolarization of the membrane at this node results in
the generation of an action potential. By this time, Na+ channels at the preceding node
are inactivated, K+ channels open, and repolarization occurs. Thus, the action potential
propagates along a myelinated axon by saltatory conduction (i.e., the jumping of an
action potential from one node to another). Myelination of an axon has two advantages:
(1) conduction is very rapid along an axon, and (2) there is a conservation of metabolic
energy because excitation is restricted to the nodal regions that are relatively small (0.5
µm).
        Conduction is also influenced by temperature, a high temperature leads to a
higher conduction velocity

       We can also note that the spinocerebellar tract has highest conduction velocity.



       26 – Electrical and chemical transmission at synapses.

       Types of Synaptic Transmission:
Two types of synaptic transmission electrical and chemical are recognized in the
nervous system. It should be noted that the electrical synapses are relatively less
common than the chemical synapses in the mammalian nervous system.

Electrical Transmission

        In electrical transmission between the nerve cells, the current generated by an
impulse in one neuron spreads to another neuron through a pathway of low electrical
resistance. Electrical synapses occur at gap junctions. In an electrical synapse, ion
channels connect the cytoplasm of the presynaptic and postsynaptic cells. In the adult
mammalian central nervous system, electrical synapses are present where the activity of
neighboring neurons needs to be highly synchronized. For example, hormone-secreting
neurons in mammalian hypothalamus are connected with electrical synapses so that they
fire almost simultaneously and secrete a burst of hormone into the circulation.
        At an electrical synapse, the current generated by voltage-gated channels at the
presynaptic neuron flows directly into the postsynaptic neuron. Therefore, transmission
at such a synapse is very rapid (<0.1 msec). At some synapses (e.g., in the giant motor
synapse of crayfish), the current can pass in one direction (from presynaptic to
postsynaptic neuron) but not in the reverse direction. Such synapses are called rectifying
or unidirectional synapses. At other synapses, the current can pass equally well in both
directions. Such synapses are called nonrectifying or bidirectional synapses. Most
electrical synapses in mammalian nervous system are believed to be the nonrectifying
type.

Chemical Transmission:

       At chemical synapses, there is no continuity between the cytoplasm of the
presynaptic terminal and postsynaptic neuron. Instead, the cells are separated by
synaptic clefts, which are fluid-filled gaps (20-50 nm). The presynaptic and
postsynaptic membranes adhere to each other due to the presence of a matrix of
extracellular fibrous protein in the synaptic cleft. The presynaptic terminal contains
synaptic vesicles that are filled with several thousand molecules of a specific chemical
substance, the neurotransmitter.
       Pyramid-like structures consisting of proteins arise from the intracellular side of
the presynaptic terminal membrane and project into the cytoplasm of the presynaptic
terminal. These pyramids and the membranes associated with them are called active
zones and are the specialized release sites in the presynaptic terminal. The vesicles
containing the neurotransmitter are aggregated near the active zones.

       Mechanisms of Transmitter Release:

       An action potential depolarizes the presynaptic nerve terminal, voltage-gated
Ca channels located in the presynaptic terminal membrane open, Ca2+ permeability
  2+

increases, and Ca2+ enters the terminal. These events cause the membrane of the
27 – Excitatory and inhibitory neurotrasmitters.


Neurotrasmitter: chemical substance that is synthesized in a neuron, released at a
synapse following depolarization of the nerve terminal (usually dependent on influx of
calcium ions), which binds to receptors on the postsynaptic cell and/or presynaptic
      terminal to elicit a specific response.
              (1) the substance must be synthesized in the neuron, and the enzymes needed for
      its synthesis must be present in the neuron;
Small Molecule it must be released in sufficient quantity to elicit a response from the
              (2)                                         Gaseous
Neurotransmittersneuron or cell located in the effector Neurotransmitters
      postsynaptic                   Neuropeptides        organ;
Acetylcholine(3) mechanisms forOpioid peptides            Nitric oxide neurotransmitter from the
                                      removal or inactivation of the
Excitatory amino acids exist; and β-endorphin,
      synaptic cleft must
  Glutamate (4) it should mimic the action of the endogenously released neurotransmitter
                                       Methionine-
  Aspartate administered exogenously at or near a synapse.
      when                           enkephalin
Inhibitory amino acids                 Leucine-enkephalin
  GABA                                 Endomorphins
  Glycine                              Nociceptin
                                                                                   Classes of
Biogenic amines                    Substance P                                     Neurotrans-
  Catecholamines                                                                   mitters
    Dopamine
    Norepinephrine
    Epinephrine
  Indoleamine
              In the CNS, a neuron is contantly bombarded by neurotrasmitters, each of which
    Serotonin (5-
      can generate or modify a synaptic potential.
hydroxytryptamine, [5-HT])
              Neurotrasmitters that move the membrane potential towars depolarization with
  Imidazole amine
      the resultant production of an action potential are known as excitatory neurotrasmitters.
    Histamine
              Neurotrasmitters that move the membrane away from depolarization by making
  Purines
      the resting membrane potential more negative, the membrane is hyperrepolariyed, are
    ATP
      known as inhibitory neurotrasmitters.
    Adenosine
              Because the postsynaptic response is actually elicited by the receptor rather than
      by the trasmitters, the postsynaptic receptor determines whether a given
      neurotransmitter will be excitatory or inhibitory. Some neurotrasmitters can have either
      effect, depending on the type of postsynaptic receptor present.

             Excitatory: Acetylcholine, glutamate, aspratate..
             Inhibitory: Dopamine, adenosine, serotonin, histamine, GABA…
             Both: Epinepherine, norepinepherine, glycine


             Acetylcholine

            Neurotransmitter in both the peripheral nervous system (PNS) and central
      nervous system (CNS). Acetylcholine is one of many neurotransmitters in the
      autonomic nervous system (ANS) and the only neurotransmitter used in the somatic
      nervous system. It is also the neurotransmitter in all autonomic ganglia.

          In the peripheral nervous system, acetylcholine activates muscles, and is a major
      neurotransmitter in the autonomic nervous system. When acetylcholine binds to
      acetylcholine receptors on skeletal muscle fibers, it opens ligand gated sodium channels
      in the cell membrane. Sodium ions then enter the muscle cell, stimulating muscle
      contraction. Acetylcholine, while inducing contraction of skeletal muscles, instead
induces decreased contraction in cardiac muscle fibers. This distinction is attributed to
differences in receptor structure between skeletal and cardiac fibers.

    In the autonomic nervous system, acetylcholine is released in the following sites:

    •   all pre- and post-ganglionic parasympathetic neurons
    •   all preganglionic sympathetic neurons
            o preganglionic sympathetic fibers to suprarenal medulla, the modified
                 sympathetic ganglion; on stimulation by acetylcholine, the suprarenal
                 medulla releases epinephrine and norepinephrine
    •   some postganglionic sympathetic fibers
            o sudomotor neurons to sweat glands.

        In the central nervous system, ACh has a variety of effects as a
neuromodulator, e.g., for plasticity and excitability. Other effects are arousal and
reward. Damage to the cholinergic system in the brain has been suggested to play a role
in the memory deficits associated with Alzheimer's Disease.




Types of acetylcholine receptors:


•   Nicotinic AChRs are ionotropic receptors permeable to sodium, potassium, and
    chloride ions. They are stimulated by nicotine and acetylcholine. They are of two
    main types, muscle type and neuronal type. The former can be selectively blocked
    by curare and the latter by hexamethonium. The main location of nicotinic AChRs is
    on muscle end plates, autonomic ganglia (both sympathetic and parasympathetic),
    and in the CNS.
•   Muscarinic receptors are metabotropic, and affect neurons over a longer time
    frame. They are stimulated by muscarine and acetylcholine, and blocked by
    atropine. Muscarinic receptors are found in both the central nervous system and the
    peripheral nervous system, in heart, lungs, upper GI tract and sweat glands.



    Glutamate:

    Some of the important physiological and clinical considerations relevant to
glutamate are as follows.

    •   Glutamate has been implicated as a transmitter in a variety of circuits in the
        brain. E.g. excitatory amino acids may be involved in learning and memory
        processes, as well as motor functions.

        Dopamine is a neurotransmitter occurring in a wide variety of animals. In the
brain, it functions as a neurotransmitter, activating the five types of dopamine receptors
— D1, D2, D3, D4 and D5, and their variants. Dopamine is produced in several areas of
the brain, including the substantia nigra and the ventral tegmental area. Dopamine is
also a neurohormone released by the hypothalamus. Its main function as a hormone is to
inhibit the release of prolactin from the anterior lobe of the pituitary.

       Epinepherine: when in the bloodstream, it rapidly prepares the body for action
in emergency situations. The hormone boosts the supply of oxygen and glucose to the
brain and muscles, while suppressing other non-emergency bodily processes. It
increases heart rate and stroke volume, dilates the pupils, and constricts arterioles in the
skin and gastrointestinal tract while dilating arterioles in skeletal muscles. It elevates the
blood sugar level by increasing catabolism of glycogen to glucose in the liver, and at the
same time begins the breakdown of lipids in fat cells. Like some other stress hormones,
epinephrine has a suppressive effect on the immune system.

    Epinephrine's actions are mediated through adrenergic receptors. Epinephrine is a
non-selective agonist of all adrenergic receptors. It activates α1, α2, β1, and β2 receptors
to different extents. Specific functions include:

   •   It binds to α1 receptors of liver cells, which activate inositol-phospholipid
       signaling pathway, signaling the phosphorylation of glycogen synthase and
       phosphorylase kinase (inactivating and activating them, respectively), leading to
       the latter activating another enzyme—glycogen phosphorylase—which catalises
       breakdown of glycogen (glycogenolysis) so as to release glucose to the
       bloodstream. Simultaneously protein phosphatase-1 (PP1) is inactivated, as in
       the active state PP1 would reverse all the previous phosphorylations.

   •   Epinephrine also activates β-adrenergic receptors of the liver and muscle cells,
       thereby activating the adenylate cyclase signaling pathway, which will in turn
       increase glycogenolysis.

    β2 receptors are found primarily in skeletal muscle blood vessels where they trigger
vasodilation. However, α-adrenergic receptors are found in most smooth muscles and
splanchnic vessels, and epinephrine triggers vasoconstriction in those vessels.

       Epinepherine is found in the spinal cord, hypothalamus, thalamus and
periaqueducts.

       Serotonin:

        Serotonin-containing cells in the raphe regions of the brainstem are believed to
play a role in descending pain-control systems. Other serotonin-containing neurons may
play a role in mediating affective processes such as aggressive behavior and arousal.
Serotonin synthesized in the pineal gland serves as a precursor for the synthesis of
melatonin, which is a neurohormone involved in regulating sleep patterns. Serotonin is
also believed to play an important role in depression.

       Histamin:

       Histamine has been implicated as a transmitter in the regulation of food and
water intake, as well as in thermoregulation and autonomic functions.
GABA:

   •   GABA is an inhibitory transmitter in many brain circuits. E.g, GABA is used as
       an inhibitory neurotransmitter by the Purkinje cells in the cerebellum. Alteration
       of GABAergic circuits has been implicated in neurological and psychiatric
       disorders like Huntington's chorea, Parkinson's disease, senile dementi…
   •   Therefore, dietary deficiency of vitamin B6 can lead to diminished GABA
       synthesis. In a disastrous series of infant deaths.. GABA content in the brain of
       these infants was reduced. Subsequently, there was a loss of synaptic inhibition
       that caused seizures and death.
   •   Since epileptic seizures can be facilitated by lack of neuronal inhibition, increase
       in the inhibitory transmitter, GABA, is helpful in terminating them
   •   Barbiturates act as agonists or modulators on postsynaptic GABA receptors and
       are used to treat epilepsy.




28 – Excitatory and inhibitory postsynaptic potentials.



        Postsynaptic potentials are changes in the membrane potential of the
postsynaptic terminal of a chemical synapse. Postsynaptic potentials are graded
potentials, and their function is to initiate or inhibit action potentials. They are caused
by the presynaptic neuron releasing neurotransmitters from the terminal button at the
end of an axon into the synaptic cleft. The neurotransmitters bind to receptors on the
postsynaptic terminal, which may be a neuron or a muscle cell in the case of a
neuromuscular junction. These are collectively referred to as postsynaptic receptors,
since they are on the membrane of the postsynaptic cell. Neurotransmitters bind to their
receptors by having a particular shape or structure, somewhat like the way a key fits
into certain locks.

       Postsynaptic potentials are subject to summation, spatially and/or temporally.

Excitatory postsynaptic potential (EPSP) is a temporary depolarization of
postsynaptic membrane potential caused by the flow of positively charged ions into the
postsynaptic cell as a result of opening of ligand-sensitive channels. A postsynaptic
potential is defined as excitatory if it makes it easier for the neuron to fire an action
potential. IPSPs are sometimes caused by an increase in positive charge outflow. The
flow of ions that causes an EPSP is an excitatory postsynaptic current (EPSC).

       Inhibitory postsynaptic postsynaptic potentials.

       An inhibitory postsynaptic potential (IPSP) is a synaptic potential that
decreases the chance that a future action potential will occur in a postsynaptic neuron or
α-motoneuron.. They can take place at all chemical synapses which utilize the secretion
of neurotransmitters to create cell to cell signaling. Inhibitory presynaptic neurons
release neurotransmitters which then bind to the postsynaptic receptors; this induces a
postsynaptic conductance change as ion channels open or close. An electrical current is
generated which changes the postsynaptic membrane potential to create a more negative
postsynaptic potential. Depolarization can also occur due to an IPSP if the reverse
potential is between the resting threshold and the action potential threshold. Another
way to look at inhibitory postsynaptic potentials is that they are also a chlorine
conductance change in the neuronal cell because it decreases the driving force.

       In general, a postsynaptic potential is dependent on the type and combination of
receptor channel, reverse potential of the postsynaptic potential, action potential
threshold voltage, ionic permeability of the ion channel, as well as the concentrations of
the ions in and out of the cell; this determines if it is excitatory or inhibitory. IPSPs
always want to keep the membrane potential more negative than the action potential
threshold and can be seen as a “transient hyperpolarization” EPSPs and IPSPs compete
with each other at numerous synapses of a neuron; this determines whether or not the
action potential at the presynaptic terminal will regenerate at the postsynaptic
membrane. Some common neurotransmitters involved in IPSPs are GABA and glycine.



29 – Describe structural arrangement of the neuromuscular junction and its
function.
The neuromuscular junction: The skeletal muscle fibers are innervated by large,
myelinated nerve fibers that originate from large motorneurons in the anterior horns of
the spinal cord. Each nerve fiber, after entering the muscle belly, normally branches and
stimulates from three to several hundred several skeletal muscle fibers. Each nereve
endings makes a junction, neuromuscular junction, with the muscle fiber near its
midpoint. The action potential initiated in the muscle fiber by the nerve signal travels in
both directions towards the muscle fiber ends.
        The nerve fibres forms a complex of branching nerve terminals that invaginate
into the surface os the muscle fiber but lie outside the muscle fiber plasma membrane.
The entire structure is called the motor end plate. It is vovered by one or more Scwmann
cells that insulate it from the surronfing fluids.
        In the junction between a single axon terminal and the muscle fiber membrane,
the invaginated membrane is called the synaptic gutter or synaptic trough, and the space
between the terminal and the fiber membrane is called the synaptic space or synaptic
cleft. This space is 20 to 30 nanometers wide. At the bottom of the gutter are numerous
smaller folds of the muscle membrane called subneural clefts, which gratly increase the
surface area at which the synaptic transmitter can act.
        In the axon terminal are many mitochondria that supply ATP, the energy source
that is used for the synthesis of an excitatory transmitter acetylcholine. The
acetylcholine in turn excites the muscle fiber membrane. Acetylcholine is synthetized in
the cytoplasm of the terminal, about 300.000 of which are normally in the terminals of a
single end plate. In the synaptic space are large quantities of the enzyme
acetylcholinesterase, which destroys acetylcholine a few milliseconds after it has been
released from the synaptic vesicles.
        When a nerve impulse reaches the neuromuscular junction, about 125 vesicles of
acetylcholine are released from the terminals into synaptic space.
On the inside surface of the neural membrane are linear dense bars. To each side
of each dense bar are protein particles that penetrate the neural membrane; these are
voltage-gated calcium channels. When an action potential spreads over the terminal,
these channels open and allow calcium ions to diffuse from the synaptic space to the
interior of the nerve terminal. The calcium ions, in turn, are believed to exert an
attractive influence on the acetylcholine vesicles, drawing them to the neural membrane
adjacent to dense bars. The vesicles then fuse with the neural membrane and empty their
acetylcholine into the synaptic space by the process of exocytosis.
Myastenia gravis and Lambert-Eaton syndrome.
        Myastenis gravis is a serious and sometimes fatal disease in which skeletal
muscles are weak. It is caused by the formation of circulatory antibodies to the muscle
type of nicotinic acetylcholine receptors. These antibodies destroy some of the receptors
and bind others to neighbouring receptors triggering their removal by endocytosis.
        Drugs affecting transmission at neuromuscular junction:
        a)         some drugs act on acetylcholine thus stimulating the muscle fiber, e.g.
                   nicotine
        b)         some drugs may block transmission e.g. muscle relaxant.
        c)         some drugs stimulate neuromuscular junction by inactivation ach-
                   acetylcholinesterase e.g. physostigmine



30 – Presynaptic inhibition and posttetanic potentiation

        In addition to the inhibition caused by inhibitory synapses operating at the
neuronal membranes which is called post-synaptic inhibition, another type of inhibition
often ocuurs in the presynaptic terminals before the signals even reaches the synapse –
pre-synaptic inhibition.
        Pre-synaptic inhibition is the phenomenon that occurs when a presynaptic
neuron exerts inhibitory influences through transmitters at an axo-axonic synapse with
the terminal in a postsynaptic neuron.
        In presynaptic inhibition, the inhibition is caused by discharge of inhibitory
synapses that lie on the presynaptic terminals nerve fibrils before their endings
terminate on the post-synaptic neuron. In most instances, the inhibitory transmitter
released is GABA, this has the specific effect of opening ion channels, allowing CL- to
diffuse into the terminal fibril.
        Therefore the action potential itself becomes greatly reduced, thus also reducing
the degree of excitation of the postsynaptic neuron.
        Presynaptic inhibition occurs in many of the sensory pathways in the nervous
system.

       3 mechanisms of presynaptic inhibition are seen:

⇒ Activation of the presynaptic receptors increases Cl- conductance, and this has been
  shown to decrease the size of the action potentials rwaching the excitatory ending.
⇒ This in turn reduces Ca++ entry and consequently the amount of excitatory
  transmitter released. Voltage-gated K+ channels are also opened and the resulting
  K+ also decreases Ca++ influx.
⇒ Evidence for direct inhibition of transmitter release independent of Ca++ influx into
  the excitatory ending is evident.
Post-tetanic potentiation

    If we stimulate motor unit and correlate it with the srenght of muscle contraction we
    can observe the principles of frequency summation and tetanization. First, with a
    lower rate of stimulation (times per second) and then increasing the frequency there
    comes a point where each new contraction occurs before the preeciding one is over.
    As a result, the second contraction is added partially to the first, so that the total
    strength of contraction rises progressively with increasing frequency. When the
    frequency reaches a critical level, the successive contractions eventually become so
    rapid that they fuse together, and the whole muscle contraction appears to be
    completely smooth and continuos. This is called tetanization. At a slightly higher
    frequency, the strength of contraction reaches its maximum, so that any additional
    increase in frequency beyond that point has no further effect in increasing
    contractile force. This occurs because enough calcium ions are maintained in the
    muscle sarcoplasm, even between action potentials, so that full contractile state is
    sustained without allowing any relaxation between the action potentials.
        The tetanizing stimulation causes Ca++ to accumulate in the presynaptic neuron
to such a degree that the intracellular binding sites that keep cytoplasm Ca++ low are
overwhelmed.
        Postetanic potentiation is opposite to habituation
31 – Spontaneously active neurons:


       Many neurons do not maintain a steady resting potential but fire impulses
spontaneously.
       Two patterns often seen are:
       a) regular firing or “beating”
       b) grouped firing or “bursting”

       In spontaneously active neurons, resting Na+ conductance is high, the leakage
current depolarizes and the threshold is crossed, and an action potential is fired.

        a) In regular firing neurons: in these neurons the membrane potential is less
negative due to their possession of high Na+ leak conductance and low K+ leak
conductancy. As the membrane potential moves to a more positive value, the thereshold
is crossed quickly. After an impulse, voltage K+ channels open and the membrane is
hyperepolarized. There is then a return to the resting potential due to to Na+ leakage.

       b)In bursting neurons: these neurons have the same membrane channels as
       firing neurons, they regulate action potentials during a burst. In depolarization of
       an action potential, voltage-gated Ca++ channels are opened to allow Ca++.
       They are slow, voltage-gated channels allow Na+, Ca++ exchange. This slow
       depolarization predominate during the impulse generating phase of the burst
       cycle. K+ channels which are activated by Ca++ are responsible for
       hyperrepolarisation (calcium-gated K+ channel).
            In bursting neurons, the leakage is slow, voltage dependent depolarizing
       channels (Na+, Ca++) generate the burst in a action potential. During this phase,
       Ca++ enters the cell by the voltage dependent Ca++ channels and activates Ca++
gated K+ channels. K + hyperrepolarizes the membrane until the moment when
       Ca++ is pumped out of the cell or sequestrated.



Spontaneous active neurons are important for circuits controlling respiration,
locomoytion as well as other activities




32 – Coding of sensory information

       Cells specialized to respond to a specific environmental stimuli are termed
sensory receptor cells.
       a) a neuron specialized for sensory reception
       b) a receptor cell connected to an afferent neuron
       c) sense organ composed of receptor cells and additional accessory to transform
           the stimulus.
       Receptor cells respond specifically to certain stimuli (receptor specificity) , there
       are 4 major groups of receptors in mammals:
       a) thermoreceptors
       b) chemoreceptors
       c) photoreceptors
       d) mechanoreceptors

       A stimulus is characterized by its modality, intensity, duration and location.
       Sensation evoked by a stimulus depends on the part of the brain that has been
       stimulated – Pacinian corpuscle stimulation either by touch or imitation from a
       tumour produces a touch sensation.
        Sensory information are processed by the thalamus and is transmitted to the
cerebral cortex where the nerve pathways from a particular sense organ are stimulated,
the sensation evoked is that for which the receptor is specialized, no matter how long
the pathway or where along the pathway the activity has been initiated.
        Law of projection: no matter where a particular sensory pathway is stimulated
along its course to the cortex, the conscious adaptation produced is referred to the
location of the receptor, (e.g. phantom limb sensation seen in amputated people or in
neurosurgical experiments with conscious patients, stimulation of specific part of cortex
may produce tickling sensations)
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2
Neuroscience sofia ultimo2

More Related Content

What's hot

Development of the nervous system
Development of the nervous systemDevelopment of the nervous system
Development of the nervous systemmuhammaduzairkhan8
 
Development of central nervous system
Development of central nervous systemDevelopment of central nervous system
Development of central nervous systemDikshya upreti
 
Protection of the cns
Protection of the cnsProtection of the cns
Protection of the cnsMichael Wrock
 
Embryology of nervous system
Embryology of nervous systemEmbryology of nervous system
Embryology of nervous systemdrnaveent
 
General histology || Nervous tissue
General histology || Nervous tissueGeneral histology || Nervous tissue
General histology || Nervous tissueAli Nasser
 
Histology slide guide
Histology slide guideHistology slide guide
Histology slide guideMUBOSScz
 
Meninges the_spinal_cord
Meninges  the_spinal_cordMeninges  the_spinal_cord
Meninges the_spinal_cordPersonal
 
Anatomy of uveal tract- ophthalmology
Anatomy of uveal tract- ophthalmologyAnatomy of uveal tract- ophthalmology
Anatomy of uveal tract- ophthalmologyNISCHAL SHRESTHA
 
Development of the musculoskeletal system
Development of the musculoskeletal systemDevelopment of the musculoskeletal system
Development of the musculoskeletal systemSahar Hafeez
 
09.22.08: Histology of the Peripheral Nervous System
09.22.08: Histology of the Peripheral Nervous System09.22.08: Histology of the Peripheral Nervous System
09.22.08: Histology of the Peripheral Nervous SystemOpen.Michigan
 

What's hot (19)

Development of the nervous system
Development of the nervous systemDevelopment of the nervous system
Development of the nervous system
 
Meninges
MeningesMeninges
Meninges
 
Development of central nervous system
Development of central nervous systemDevelopment of central nervous system
Development of central nervous system
 
Protection of the cns
Protection of the cnsProtection of the cns
Protection of the cns
 
Anatomy of uvea
Anatomy of uveaAnatomy of uvea
Anatomy of uvea
 
Embryology of nervous system
Embryology of nervous systemEmbryology of nervous system
Embryology of nervous system
 
Neuroglia
NeurogliaNeuroglia
Neuroglia
 
Meninges - Anatomy of the Meninges & CSF
Meninges - Anatomy of the Meninges & CSFMeninges - Anatomy of the Meninges & CSF
Meninges - Anatomy of the Meninges & CSF
 
General histology || Nervous tissue
General histology || Nervous tissueGeneral histology || Nervous tissue
General histology || Nervous tissue
 
Histology slide guide
Histology slide guideHistology slide guide
Histology slide guide
 
Nervous tissue histology
Nervous tissue  histology Nervous tissue  histology
Nervous tissue histology
 
Meninges the_spinal_cord
Meninges  the_spinal_cordMeninges  the_spinal_cord
Meninges the_spinal_cord
 
Ciliary body
Ciliary bodyCiliary body
Ciliary body
 
Anatomy of uveal tract- ophthalmology
Anatomy of uveal tract- ophthalmologyAnatomy of uveal tract- ophthalmology
Anatomy of uveal tract- ophthalmology
 
Development of the musculoskeletal system
Development of the musculoskeletal systemDevelopment of the musculoskeletal system
Development of the musculoskeletal system
 
09.22.08: Histology of the Peripheral Nervous System
09.22.08: Histology of the Peripheral Nervous System09.22.08: Histology of the Peripheral Nervous System
09.22.08: Histology of the Peripheral Nervous System
 
Meninges with it's clinical aspects
Meninges with it's clinical aspectsMeninges with it's clinical aspects
Meninges with it's clinical aspects
 
Anatomy of Sclera
Anatomy of ScleraAnatomy of Sclera
Anatomy of Sclera
 
Meninges and csf
Meninges and csfMeninges and csf
Meninges and csf
 

Viewers also liked

07 derivatives ii
07 derivatives ii07 derivatives ii
07 derivatives iiMUBOSScz
 
Cranial nerves
Cranial nervesCranial nerves
Cranial nervesMUBOSScz
 
Tooth, esophagus, stomach, small intestine
Tooth, esophagus, stomach, small intestineTooth, esophagus, stomach, small intestine
Tooth, esophagus, stomach, small intestineMUBOSScz
 
Captain’s role
Captain’s roleCaptain’s role
Captain’s roleMUBOSScz
 
05 elements _ros__org._comp
05 elements _ros__org._comp05 elements _ros__org._comp
05 elements _ros__org._compMUBOSScz
 
0 introductory recapitulation
0 introductory recapitulation0 introductory recapitulation
0 introductory recapitulationMUBOSScz
 
Electrochemanal labauxiliary-fin
Electrochemanal labauxiliary-finElectrochemanal labauxiliary-fin
Electrochemanal labauxiliary-finMUBOSScz
 
14 proteins
14 proteins14 proteins
14 proteinsMUBOSScz
 
04 redox reactions__dissoln.__precip
04 redox reactions__dissoln.__precip04 redox reactions__dissoln.__precip
04 redox reactions__dissoln.__precipMUBOSScz
 
06 nomenclature _hydrocarbons__deriv._i
06 nomenclature _hydrocarbons__deriv._i06 nomenclature _hydrocarbons__deriv._i
06 nomenclature _hydrocarbons__deriv._iMUBOSScz
 
02 hydrolysis. buffers__colloids
02 hydrolysis. buffers__colloids02 hydrolysis. buffers__colloids
02 hydrolysis. buffers__colloidsMUBOSScz
 
15 glycoproteins _haemoproteins
15 glycoproteins _haemoproteins15 glycoproteins _haemoproteins
15 glycoproteins _haemoproteinsMUBOSScz
 
10 polysacch. _heteroglycosides__nucleic_acids
10 polysacch. _heteroglycosides__nucleic_acids10 polysacch. _heteroglycosides__nucleic_acids
10 polysacch. _heteroglycosides__nucleic_acidsMUBOSScz
 
03 kinetics _energetics
03 kinetics _energetics03 kinetics _energetics
03 kinetics _energeticsMUBOSScz
 
01 solutions _electrolytes__protolytic_equilibria
01 solutions _electrolytes__protolytic_equilibria01 solutions _electrolytes__protolytic_equilibria
01 solutions _electrolytes__protolytic_equilibriaMUBOSScz
 
11 lipids i_-_fa__simple_and_complex_lipids
11 lipids i_-_fa__simple_and_complex_lipids11 lipids i_-_fa__simple_and_complex_lipids
11 lipids i_-_fa__simple_and_complex_lipidsMUBOSScz
 
Lymfomyobrne
LymfomyobrneLymfomyobrne
LymfomyobrneMUBOSScz
 
12 lipids ii_-_eicosanoids__isoprenoids__steroids
12 lipids ii_-_eicosanoids__isoprenoids__steroids12 lipids ii_-_eicosanoids__isoprenoids__steroids
12 lipids ii_-_eicosanoids__isoprenoids__steroidsMUBOSScz
 

Viewers also liked (20)

07 derivatives ii
07 derivatives ii07 derivatives ii
07 derivatives ii
 
Cranial nerves
Cranial nervesCranial nerves
Cranial nerves
 
Tooth, esophagus, stomach, small intestine
Tooth, esophagus, stomach, small intestineTooth, esophagus, stomach, small intestine
Tooth, esophagus, stomach, small intestine
 
Captain’s role
Captain’s roleCaptain’s role
Captain’s role
 
Cz uk
Cz ukCz uk
Cz uk
 
05 elements _ros__org._comp
05 elements _ros__org._comp05 elements _ros__org._comp
05 elements _ros__org._comp
 
0 introductory recapitulation
0 introductory recapitulation0 introductory recapitulation
0 introductory recapitulation
 
Electrochemanal labauxiliary-fin
Electrochemanal labauxiliary-finElectrochemanal labauxiliary-fin
Electrochemanal labauxiliary-fin
 
14 proteins
14 proteins14 proteins
14 proteins
 
04 redox reactions__dissoln.__precip
04 redox reactions__dissoln.__precip04 redox reactions__dissoln.__precip
04 redox reactions__dissoln.__precip
 
06 nomenclature _hydrocarbons__deriv._i
06 nomenclature _hydrocarbons__deriv._i06 nomenclature _hydrocarbons__deriv._i
06 nomenclature _hydrocarbons__deriv._i
 
Pregn
PregnPregn
Pregn
 
02 hydrolysis. buffers__colloids
02 hydrolysis. buffers__colloids02 hydrolysis. buffers__colloids
02 hydrolysis. buffers__colloids
 
15 glycoproteins _haemoproteins
15 glycoproteins _haemoproteins15 glycoproteins _haemoproteins
15 glycoproteins _haemoproteins
 
10 polysacch. _heteroglycosides__nucleic_acids
10 polysacch. _heteroglycosides__nucleic_acids10 polysacch. _heteroglycosides__nucleic_acids
10 polysacch. _heteroglycosides__nucleic_acids
 
03 kinetics _energetics
03 kinetics _energetics03 kinetics _energetics
03 kinetics _energetics
 
01 solutions _electrolytes__protolytic_equilibria
01 solutions _electrolytes__protolytic_equilibria01 solutions _electrolytes__protolytic_equilibria
01 solutions _electrolytes__protolytic_equilibria
 
11 lipids i_-_fa__simple_and_complex_lipids
11 lipids i_-_fa__simple_and_complex_lipids11 lipids i_-_fa__simple_and_complex_lipids
11 lipids i_-_fa__simple_and_complex_lipids
 
Lymfomyobrne
LymfomyobrneLymfomyobrne
Lymfomyobrne
 
12 lipids ii_-_eicosanoids__isoprenoids__steroids
12 lipids ii_-_eicosanoids__isoprenoids__steroids12 lipids ii_-_eicosanoids__isoprenoids__steroids
12 lipids ii_-_eicosanoids__isoprenoids__steroids
 

Similar to Neuroscience sofia ultimo2

Anatomi fisiologi saraf (neuron)
Anatomi fisiologi saraf (neuron)Anatomi fisiologi saraf (neuron)
Anatomi fisiologi saraf (neuron)DwiKartikaRukmi
 
Neurons and the glia.pptx
Neurons and the glia.pptxNeurons and the glia.pptx
Neurons and the glia.pptxSabaJahan2
 
Introduction to neurons
Introduction to neuronsIntroduction to neurons
Introduction to neuronsmahnoorIjaz6
 
5.1. Nerve Tissue physiology.pptx
5.1. Nerve Tissue physiology.pptx5.1. Nerve Tissue physiology.pptx
5.1. Nerve Tissue physiology.pptxMonenusKedir
 
Neurotransmission and neuromuscular junction
Neurotransmission and neuromuscular junctionNeurotransmission and neuromuscular junction
Neurotransmission and neuromuscular junctionInbarajAnandan
 
Nerve tissues (mic ana lec)
Nerve tissues (mic ana lec)Nerve tissues (mic ana lec)
Nerve tissues (mic ana lec)ayesexy
 
Comparing Two Different Types Of Neurons
Comparing Two Different Types Of NeuronsComparing Two Different Types Of Neurons
Comparing Two Different Types Of NeuronsNatasha Barnett
 
Cellular organization of the nervous system
Cellular organization of the nervous systemCellular organization of the nervous system
Cellular organization of the nervous systemDavis Mburu
 
Chap_9_Control__Coordination_(2).pptx
Chap_9_Control__Coordination_(2).pptxChap_9_Control__Coordination_(2).pptx
Chap_9_Control__Coordination_(2).pptxRahul Badve
 
Lect. 9 nervous tissues
Lect. 9   nervous tissuesLect. 9   nervous tissues
Lect. 9 nervous tissuesHara O.
 
Cells of Nervous System
Cells of Nervous SystemCells of Nervous System
Cells of Nervous Systemvacagodx
 
Neurophysiology1
Neurophysiology1Neurophysiology1
Neurophysiology1Essy Essy
 
Histology of nervous tissue1
Histology of nervous tissue1Histology of nervous tissue1
Histology of nervous tissue1Kifayat Khan
 
Structure of neuron & synapse
Structure of neuron & synapse Structure of neuron & synapse
Structure of neuron & synapse Hridya Prasad
 
The growth and guidance of axons in body.pptx
The growth and guidance of axons in body.pptxThe growth and guidance of axons in body.pptx
The growth and guidance of axons in body.pptxNitish kumar
 

Similar to Neuroscience sofia ultimo2 (20)

Anatomi fisiologi saraf (neuron)
Anatomi fisiologi saraf (neuron)Anatomi fisiologi saraf (neuron)
Anatomi fisiologi saraf (neuron)
 
Neurons and the glia.pptx
Neurons and the glia.pptxNeurons and the glia.pptx
Neurons and the glia.pptx
 
Nerve cell
Nerve cellNerve cell
Nerve cell
 
Introduction to neurons
Introduction to neuronsIntroduction to neurons
Introduction to neurons
 
5.1. Nerve Tissue physiology.pptx
5.1. Nerve Tissue physiology.pptx5.1. Nerve Tissue physiology.pptx
5.1. Nerve Tissue physiology.pptx
 
Neurotransmission and neuromuscular junction
Neurotransmission and neuromuscular junctionNeurotransmission and neuromuscular junction
Neurotransmission and neuromuscular junction
 
Nerve tissues (mic ana lec)
Nerve tissues (mic ana lec)Nerve tissues (mic ana lec)
Nerve tissues (mic ana lec)
 
Comparing Two Different Types Of Neurons
Comparing Two Different Types Of NeuronsComparing Two Different Types Of Neurons
Comparing Two Different Types Of Neurons
 
Cellular organization of the nervous system
Cellular organization of the nervous systemCellular organization of the nervous system
Cellular organization of the nervous system
 
Nervous System
Nervous SystemNervous System
Nervous System
 
Nueron ppt
Nueron pptNueron ppt
Nueron ppt
 
Chap_9_Control__Coordination_(2).pptx
Chap_9_Control__Coordination_(2).pptxChap_9_Control__Coordination_(2).pptx
Chap_9_Control__Coordination_(2).pptx
 
Nervous tissue
Nervous tissueNervous tissue
Nervous tissue
 
Lect. 9 nervous tissues
Lect. 9   nervous tissuesLect. 9   nervous tissues
Lect. 9 nervous tissues
 
Cells of Nervous System
Cells of Nervous SystemCells of Nervous System
Cells of Nervous System
 
Neurons
NeuronsNeurons
Neurons
 
Neurophysiology1
Neurophysiology1Neurophysiology1
Neurophysiology1
 
Histology of nervous tissue1
Histology of nervous tissue1Histology of nervous tissue1
Histology of nervous tissue1
 
Structure of neuron & synapse
Structure of neuron & synapse Structure of neuron & synapse
Structure of neuron & synapse
 
The growth and guidance of axons in body.pptx
The growth and guidance of axons in body.pptxThe growth and guidance of axons in body.pptx
The growth and guidance of axons in body.pptx
 

More from MUBOSScz

BIOCHEMISTRY II EXAM ANSWERS
BIOCHEMISTRY II EXAM ANSWERSBIOCHEMISTRY II EXAM ANSWERS
BIOCHEMISTRY II EXAM ANSWERSMUBOSScz
 
Respiratory syst copy
Respiratory syst   copyRespiratory syst   copy
Respiratory syst copyMUBOSScz
 
Practicals 3 digestive system iii
Practicals 3   digestive system iiiPracticals 3   digestive system iii
Practicals 3 digestive system iiiMUBOSScz
 
Epithelium copy
Epithelium   copyEpithelium   copy
Epithelium copyMUBOSScz
 
Cytology copy
Cytology   copyCytology   copy
Cytology copyMUBOSScz
 
Connective tissue proper copy
Connective tissue proper   copyConnective tissue proper   copy
Connective tissue proper copyMUBOSScz
 
Cartilage, bone copy
Cartilage, bone   copyCartilage, bone   copy
Cartilage, bone copyMUBOSScz
 
Cardiovascular system copy
Cardiovascular system   copyCardiovascular system   copy
Cardiovascular system copyMUBOSScz
 
Bone, cartilage copy
Bone, cartilage   copyBone, cartilage   copy
Bone, cartilage copyMUBOSScz
 
Blood development copy
Blood development   copyBlood development   copy
Blood development copyMUBOSScz
 
Tissue processing
Tissue processingTissue processing
Tissue processingMUBOSScz
 
Section a dermatology
Section a dermatologySection a dermatology
Section a dermatologyMUBOSScz
 
Oncology section a
Oncology section aOncology section a
Oncology section aMUBOSScz
 
Section b dermatology
Section b dermatologySection b dermatology
Section b dermatologyMUBOSScz
 
Working and training in the national health service a guide for im gs final
Working and training in the national health service   a guide for im gs finalWorking and training in the national health service   a guide for im gs final
Working and training in the national health service a guide for im gs finalMUBOSScz
 
Inflammation
InflammationInflammation
InflammationMUBOSScz
 
Infectious diseases20
Infectious diseases20Infectious diseases20
Infectious diseases20MUBOSScz
 

More from MUBOSScz (20)

BIOCHEMISTRY II EXAM ANSWERS
BIOCHEMISTRY II EXAM ANSWERSBIOCHEMISTRY II EXAM ANSWERS
BIOCHEMISTRY II EXAM ANSWERS
 
Respiratory syst copy
Respiratory syst   copyRespiratory syst   copy
Respiratory syst copy
 
Practicals 3 digestive system iii
Practicals 3   digestive system iiiPracticals 3   digestive system iii
Practicals 3 digestive system iii
 
Epithelium copy
Epithelium   copyEpithelium   copy
Epithelium copy
 
Cytology copy
Cytology   copyCytology   copy
Cytology copy
 
Connective tissue proper copy
Connective tissue proper   copyConnective tissue proper   copy
Connective tissue proper copy
 
Cartilage, bone copy
Cartilage, bone   copyCartilage, bone   copy
Cartilage, bone copy
 
Cardiovascular system copy
Cardiovascular system   copyCardiovascular system   copy
Cardiovascular system copy
 
Bone, cartilage copy
Bone, cartilage   copyBone, cartilage   copy
Bone, cartilage copy
 
Blood development copy
Blood development   copyBlood development   copy
Blood development copy
 
Tissue processing
Tissue processingTissue processing
Tissue processing
 
Section a dermatology
Section a dermatologySection a dermatology
Section a dermatology
 
Oncology section a
Oncology section aOncology section a
Oncology section a
 
Section b dermatology
Section b dermatologySection b dermatology
Section b dermatology
 
Working and training in the national health service a guide for im gs final
Working and training in the national health service   a guide for im gs finalWorking and training in the national health service   a guide for im gs final
Working and training in the national health service a guide for im gs final
 
Preparaty
PreparatyPreparaty
Preparaty
 
Perfusion
PerfusionPerfusion
Perfusion
 
Pedi en
Pedi enPedi en
Pedi en
 
Inflammation
InflammationInflammation
Inflammation
 
Infectious diseases20
Infectious diseases20Infectious diseases20
Infectious diseases20
 

Recently uploaded

My INSURER PTE LTD - Insurtech Innovation Award 2024
My INSURER PTE LTD - Insurtech Innovation Award 2024My INSURER PTE LTD - Insurtech Innovation Award 2024
My INSURER PTE LTD - Insurtech Innovation Award 2024The Digital Insurer
 
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)Mark Simos
 
Benefits Of Flutter Compared To Other Frameworks
Benefits Of Flutter Compared To Other FrameworksBenefits Of Flutter Compared To Other Frameworks
Benefits Of Flutter Compared To Other FrameworksSoftradix Technologies
 
Beyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry InnovationBeyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry InnovationSafe Software
 
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 3652toLead Limited
 
Scanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsScanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsRizwan Syed
 
Key Features Of Token Development (1).pptx
Key  Features Of Token  Development (1).pptxKey  Features Of Token  Development (1).pptx
Key Features Of Token Development (1).pptxLBM Solutions
 
Pigging Solutions Piggable Sweeping Elbows
Pigging Solutions Piggable Sweeping ElbowsPigging Solutions Piggable Sweeping Elbows
Pigging Solutions Piggable Sweeping ElbowsPigging Solutions
 
My Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 PresentationMy Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 PresentationRidwan Fadjar
 
"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr Bagan"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr BaganFwdays
 
costume and set research powerpoint presentation
costume and set research powerpoint presentationcostume and set research powerpoint presentation
costume and set research powerpoint presentationphoebematthew05
 
Dev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio WebDev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio WebUiPathCommunity
 
AI as an Interface for Commercial Buildings
AI as an Interface for Commercial BuildingsAI as an Interface for Commercial Buildings
AI as an Interface for Commercial BuildingsMemoori
 
Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!Commit University
 
Install Stable Diffusion in windows machine
Install Stable Diffusion in windows machineInstall Stable Diffusion in windows machine
Install Stable Diffusion in windows machinePadma Pradeep
 
Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?Mattias Andersson
 
Unleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding ClubUnleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding ClubKalema Edgar
 
Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024BookNet Canada
 

Recently uploaded (20)

My INSURER PTE LTD - Insurtech Innovation Award 2024
My INSURER PTE LTD - Insurtech Innovation Award 2024My INSURER PTE LTD - Insurtech Innovation Award 2024
My INSURER PTE LTD - Insurtech Innovation Award 2024
 
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
Tampa BSides - Chef's Tour of Microsoft Security Adoption Framework (SAF)
 
Benefits Of Flutter Compared To Other Frameworks
Benefits Of Flutter Compared To Other FrameworksBenefits Of Flutter Compared To Other Frameworks
Benefits Of Flutter Compared To Other Frameworks
 
Beyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry InnovationBeyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
Beyond Boundaries: Leveraging No-Code Solutions for Industry Innovation
 
Vulnerability_Management_GRC_by Sohang Sengupta.pptx
Vulnerability_Management_GRC_by Sohang Sengupta.pptxVulnerability_Management_GRC_by Sohang Sengupta.pptx
Vulnerability_Management_GRC_by Sohang Sengupta.pptx
 
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
Tech-Forward - Achieving Business Readiness For Copilot in Microsoft 365
 
Scanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL CertsScanning the Internet for External Cloud Exposures via SSL Certs
Scanning the Internet for External Cloud Exposures via SSL Certs
 
Key Features Of Token Development (1).pptx
Key  Features Of Token  Development (1).pptxKey  Features Of Token  Development (1).pptx
Key Features Of Token Development (1).pptx
 
Pigging Solutions Piggable Sweeping Elbows
Pigging Solutions Piggable Sweeping ElbowsPigging Solutions Piggable Sweeping Elbows
Pigging Solutions Piggable Sweeping Elbows
 
My Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 PresentationMy Hashitalk Indonesia April 2024 Presentation
My Hashitalk Indonesia April 2024 Presentation
 
"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr Bagan"ML in Production",Oleksandr Bagan
"ML in Production",Oleksandr Bagan
 
costume and set research powerpoint presentation
costume and set research powerpoint presentationcostume and set research powerpoint presentation
costume and set research powerpoint presentation
 
Dev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio WebDev Dives: Streamline document processing with UiPath Studio Web
Dev Dives: Streamline document processing with UiPath Studio Web
 
AI as an Interface for Commercial Buildings
AI as an Interface for Commercial BuildingsAI as an Interface for Commercial Buildings
AI as an Interface for Commercial Buildings
 
Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!Nell’iperspazio con Rocket: il Framework Web di Rust!
Nell’iperspazio con Rocket: il Framework Web di Rust!
 
Install Stable Diffusion in windows machine
Install Stable Diffusion in windows machineInstall Stable Diffusion in windows machine
Install Stable Diffusion in windows machine
 
Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?Are Multi-Cloud and Serverless Good or Bad?
Are Multi-Cloud and Serverless Good or Bad?
 
E-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptx
E-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptxE-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptx
E-Vehicle_Hacking_by_Parul Sharma_null_owasp.pptx
 
Unleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding ClubUnleash Your Potential - Namagunga Girls Coding Club
Unleash Your Potential - Namagunga Girls Coding Club
 
Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024
Transcript: New from BookNet Canada for 2024: BNC BiblioShare - Tech Forum 2024
 

Neuroscience sofia ultimo2

  • 1. 1-Cytoskeletal components of the neurons and their functions during axon regeneration. Neurons contain a cytoskeleton consisting of neurofibrils, which determine the shape of the soma and the various processes extending from it, and which transport substances throught the neuron Microtubules and microtubule associated proteins (MAP)  25-28nm  are narrow longitudinal tubes present in all neuronal processes. The tubesmantain shape, and also transport molecules such as neuro-transmitters from the soma to the axon terminals (anterograd transport), or from the terminals to the soma (retrograde transport). There are at least two types of axonal transport: (a) rapid, 400nm (b) slow, at less than 1 mm day Microfilaments  5-7 nm  present in high concentrations as a meshwork beneath the membrane of the axon. Actin is a important protein in axon development , and causes the movement of the growth cone.  Are a proliferation of globulin actin (G-actin) in the axolemma of the synapse, depolarisation of the microfilaments is a prerequesite for releasing of the trasmitters from the synapse vesicles. Neurofilamets  10nm  Are the most abundant of the fibrillar elements in the neuron, and form the bones of the cytoskeleton  they are largely spaced, having tide arms that keep them appart and provide neuronal stability  seem to be invoolved in the transport mechanisms 3 different types of neurons: 1. unipolar 2. bipolar – concentrated mainly in periphery 3. multi-polar – formed by alfa and beta tubules according to size: 1. Type I Golgi (large+long axons) 2. Type II Golgi (small+short axons)
  • 2. 2-Types of axoplasmatic transports according direction and speed, their functions in the intact neuron and during axon regeneration. Axonal Transport: Various secretory products produced in the cell body are carried to the axon terminals by special transport mechanism as in the same manner various constituents are carried from the axon terminals to the cell body. Three main types of axonal transport are: ⇒ fast anterograde transport ⇒ slow anterograde transport ⇒ fast retrograde transport. Fast anterograde transport is involved in the transport of materials that have a functional role at the nerve terminals (e.g., precursors of peptide neurotransmitters, enzymes needed for the synthesis of small molecule neurotransmitters, and glycoproteins needed for reconstitution of the plasma membrane) are trasported from the cell body to the terminals. Polypeptides much larger than final peptide neurotransmitters (pre-propeptides) and enzymes needed for the synthesis of small molecule neurotransmitters are synthesized in the rough endoplasmic reticulum. The vesicles formed in the Golgi apparatus for the axon terminals then become attached to the microtubules and are transported by fast axonal transport (at a rate of 400 mm/d) into the nerve terminal. The rapid axonal transport depends on the microtubules. The microtubule provides a stationary track and a microtubule-associated ATPase (kinesin) forms a cross-bridge between the organelle to be moved and the microtubule. On one end, kinesin contains two globular heads that bind to the microtubule, and on the other end, it has a fan-shaped tail that binds to the surface of an organelle. The organelle then moves by sliding of the kinesin molecule along the microtubule Slow anterograde transport involves movement of neurofilaments and microtubules synthesized in the cell body to the terminals at a rate of 5 mm/d. Soluble proteins transported by this mechanism include actin, tubulin (which polymerizes to form microtubules), proteins that make up neurofilaments, myosin, and a calcium-binding protein (calmodulin). Fast retrograde transport is slower than the fast anterograde transport (about 200 mm/ d). Rapid retrograde transport carries materials from the nerve terminals to the cell body; the transported materials travel along microtubules. An example of transport by this mechanism is nerve growth factor (NGF), a peptide synthesized by a target cell and transported into certain neurons in order to stimulate their growth. Materials lying outside the axon terminals are taken up by endocytosis and transported to the cell body. Fast retrograde axonal transport is also involved in some pathological conditions. For example, the herpes simplex, polio, and rabies viruses and tetanus toxin are taken up by the axon terminals in peripheral nerves and carried to their cell bodies in the central nervous system (CNS) by rapid retrograde transport.
  • 3. 3-Types of the neuronal synapses, their classification according morphology and neurotransmitters. Synapses needed for communication e.g.interneuronal synapses or intermuscular Direct synapses in neuromuscular junction Indirect synapses in autonomic postganglionic synapse Synapses may be either: 1. interneuronal connections 2. connections between the neuron and effector Interneuronal connections according to position of connections: - axodendritic and axo somatic synapses - axo-axonal and dendro – dendritic synapses according to effect on postsynaptic element -excitatory synapses -inhibitory synapses according to transmission of the excitation - eletric synapses: - reduced extracellular space - transport of info by ionic flow -continuity of cytoplasm -chemical synapses: -presynaptic termination - postsynaptic element - active zone - synaptic cleft – no continuity between pre and post- synapses -increased extracellular space
  • 4. - transport of info by neurotransmitters 4-Glial cell types and their participation in the ontogenetic development of the CNS The supporting cells located in the CNS are called neuroglia or simply glial cells. They are nonexcitable and more numerous (5 to 10 times) than neurons. Neuroglia have been classified into the following groups: astrocytes, oligodendrocytes, microglia, and ependymal cells. ⇒ Astrocytes Among the glial cells, astrocytes are the largest and have a stellate (star-shaped) appearance because their processes extend in all directions. Their nuclei are ovoid and centrally located. The astrocytes provide support for the neurons, a barrier against the spread of transmitters from synapses, and insulation to prevent electrical activity of one neuron from affecting the activity of a neighboring neuron. They are further subdivided into the following subgroups: a) Protoplasmic Astrocytes: These cells are present in the gray matter in close association with neurons. Because of their close association with the neurons, they are considered satellite cells and serve as metabolic intermediaries for neurons. They give out thicker and shorter processes, which branch profusely. Several of their processes terminate in expansions called end-feet. Abutting of processes of protoplasmic astrocytes on the capillaries as perivascular end-feet is one of the anatomical features of the blood-brain barrier b) Fibrous Astrocytes These glial cells are found primarily in the white matter between nerve fibers. Several thin, long, and smooth processes arise from the cell body; these processes show little branching. Fibrous astrocytes function to repair damaged tissue, and this process may result in scar formation. c) MĂĽller Cells These modified astrocytes are present in the retina.
  • 5. Oligodendrocytes These cells are smaller than astrocytes and have fewer and shorter branches. Their cytoplasm contains the usual organelles (e.g., ribosomes, mitochondria, and microtubules), but they do not contain neurofilaments. In the white matter, oligodendrocytes are located in rows along myelinated fibers and are known as interfascicular oligodendrocytes. These oligodendrocytes are involved in the myelination process. The oligodendrocytes present in the gray matter are called perineural oligodendrocytes. ⇒ Microglia These are the smallest of the glial cells. They usually have a few short branching processes with thorn-like endings. These processes arising from the cell body give off numerous spine-like projections. They are scattered throughout the nervous system. When the CNS is injured, the microglia become enlarged, mobile, and phagocytic. ⇒ Ependymal Cells Ependymal cells consist of three types of cells: a) Ependymocytes are cuboidal or columnar cells that form a single layer of lining in the brain ventricles and the central canal of the spinal cord. They possess microvilli and cilia. The presence of microvilli indicates that these cells may have some absorptive function. The movement of their cilia facilitates the flow of the cerebrospinal fluid. b) Tanycytes are specialized ependymal cells that are found in the floor of the third ventricle, and their processes extend into the brain tissue where they are juxtaposed to blood vessels and neurons. Tanycytes have been implicated in the transport of hormones from the CSF to capillaries of portal system and from hypothalamic neurons to the CSF.
  • 6. c) Choroidal epithelial cells are modified ependymal cells. They are present in the choroid plexus and are involved in the production and secretion of CSF. They have tight junctions that prevent the CSF from spreading to the adjacent tissues ⇒ PNS In the peripheral nervous system (PNS), Schwann cells provide myelin sheaths around axons. The myelin sheaths are interrupted along the length of the axons at regular intervals at the nodes of Ranvier. Thus, the nodes of Ranvier are + uninsulated and have a lower resistance. These nodes of Ranvier are rich in Na channels, and the action potential becomes regenerated at these regions. Therefore, the action potential traveling along the length of the axon jumps from one node of Ranvier to another. This type of propagation enables the action potential to conduct rapidly and is known as saltatory conduction. During the myelination, the axon comes in contact with the Schwann cell, which then rotates around the axon in clockwise or counterclockwise fashion. As the Schwann cell wraps around the axon, the cytoplasm becomes progressively reduced, and the inner layers of the plasma membrane come in contact and fuse together. ⇒ CNS Within the brain and the spinal cord, oligodendrocytes form the myelin sheaths around axons of neurons. Several glial processes arise from one oligodendrocyte and wrap around a portion of the axon. The intervals between adjacent oligodendrocytes are devoid of myelin sheaths and are called the nodes of Ranvier. Unlike in peripheral axons, the process of an oligodendrocyte does not rotate spirally on the axon. Instead, it may wrap around the length of the axon. The cytoplasm is reduced progressively, and the sheath consists of concentric layers of plasma membrane. Unlike in peripheral nerves, one oligodendrocyte forms myelin sheaths around numerous (as many as 60) axons of diverse origins.
  • 7. 5-Glial cell types and their involvement in the ontogenetic development of the PNS.  Glial cells of PNS originate from neural crest cells (plate)  Schwmann cells produce myelin sheaths around myelinated axons of PNS neurons.  Glial cells provide support as well as protection for neurons  Most glial derived from ectodermal tissue (particularly neural tube and crest)  The exception is microglia- derive from mesoderm .  sattelite cells surrond neuronal cell bodies in PNS  Schwmann cells of PNS promote regeneration of peripheral neurons A myelinated nerve fiber is one that is surronded by a myelin sheath. In the CNS the supporting cell is called oligodendrocytes, in the PNS is called Schmann cell. The development of the myelin sheath provides an indication of the construction of its lamellae. The body of Schmann cell forms a rolled up sheet of paper in which the axon become embebedded. The structure develops, its margins become approximated and eventually meet together, which results in duplication of the cell membrane – the mesaxon (pair of parallel plasma membranes of a Schwann cell, marking the point of edge-to-edge contact by the Schwann cell encircling the axon). This becomes spirally bound around the axon, probably big movement of the Scwmann cell around the enclosed axon. The beginning of the duplication lies on the inner side of the myelin sheath (inner mesaxon) and its end on the outer side (outer mesaxon). Another type of supporting cell are the satellite cells. Both Schwmann cellas and satellite cells develop from neural crest cells. Myelinating Schwann cells begin to form the myelin sheath in mammals during fetal development and work by spiraling around the axon, sometimes with as many as 100 revolutions. A well-developed Schwann cell is shaped like a rolled-up sheet of paper, with layers of myelin in between each coil. The inner layers of the wrapping, which are predominantly membrane material, form the myelin sheath while the outermost layer of nucleated cytoplasm forms the neurolemma. Only a small volume of residual cytoplasm communicates the inner from the outer layers. This is seen histologically as the Schmidt-Lantermann Incisure. Since each Schwann cell can cover
  • 8. about a millimeter (0.04 inches) along the axon, hundreds and often thousands are needed to completely cover an axon, which can sometimes span the length of a body. 6-Describe Wallerian degeneration and different reactions of the glial cells in CNS and PNS following injury. ⇒ Neuronal Injury/ Injury of the Neuronal Cell Body The neuronal cell body may be damaged by disease, ischemia (lack of blood supply), or trauma. In the CNS (the brain and spinal cord), the debris produced by neuronal damage is phagocytosed by microglia. The adjacent fibrous astrocytes proliferate, and the neurons are replaced by scar tissue. In the PNS, macrophages are responsible for the removal of the debris produced by neuronal damage, and the scar tissue is produced by the proliferation of the fibroblasts. Necrotic cell death is caused by acute traumatic injury that involves rapid lysis of cell membranes. Necrotic cell death is different from apoptosis. Apoptosis is defined as a genetically determined process of cell death and is characterized by shrinkage of the cell, cellular fragmentation, and condensation of the chromatin. During the process of formation of tissues from undifferentiated germinal cells in the embryo (histogenesis), more neurons (about 2 times more) are formed than the neurons present in the mature brain. The excess number of neurons is destroyed during the development by apoptosis. The mechanism of apoptosis involves activation of a latent biochemical pathway that is present in neurons and other cells of the body. The cellular debris after neuronal cell death is removed by phagocytosis, which involves transport of solid material into the cells (e.g., microglia) that remove the debris by indentation of the cell membrane of the phagocyte and formation of a vesicle. Pinocytosis is similar to phagocytosis, except that liquid material is removed. Exocytosis involves fusion of a vesicle inside the nerve terminal (e.g., a vesicle containing a neurotransmitter) with the plasma membrane and transportation of the contents of the vesicle outside the nerve terminal.
  • 9. Axonal Damage/ Wallerian Degeneration This type of degeneration refers to the changes that occur distally to the site of damage on an axon. Because protein synthesis occurs primarily in the neuronal cell body, the segment distal to the damaged site on the axon is affected profoundly. Initially, the axon swells up and becomes irregular. Later, the axon and the terminal are broken down into fragments that are phagocytosed by adjacent macrophages and Schwann cells. Myelin is converted into fine drops of lipid material in the Schwann cells and is extruded from these cells; it is removed by macrophages in the PNS and microglial cells and invading macrophages in the CNS. Alterations may also be present in the proximal segment of the axon up to the first node of Ranvier. ⇒ Chromatolysis Sectioning of an axon may produce changes in the cell body, and if the injury is close to the cell body, the neuron may degenerate. The cell body swells up due to edema and becomes round in appearance, and the Nissl substance gets distributed throughout the cytoplasm. The nucleus moves from its central position to the periphery due to edema. The degenerative changes start within hours and are complete within a relatively short time (about a week). ⇒ Anterograde Transneuronal Degeneration This type of degeneration occurs in the CNS when damage to a neuron results in the degeneration of another postsynaptic neuron closely associated with the same function. For example, damage to an optic nerve results in the degeneration of the lateral geniculate neurons receiving inputs from this nerve. ⇒ Retrograde Transneuronal Degeneration This type of degeneration occurs in neurons sending inputs to an injured neuron. In this situation, terminals of the neuron synapsing with a chromatolytic neuron withdraw and are replaced by processes of glial cells. The neuron, from which the inputs to the chromatolytic neuron arise, eventually degenerates.
  • 10. Recovery of Neuronal Injury (Regeneration) If the damage to the neurons is not severe and they survive the injury, regeneration is possible, but complete recovery may take as long as 3 to 6 months. Within about 3 weeks, the swelling of the cell subsides, the nucleus occupies a central position in the cell body again, and the Nissl bodies are normally distinguished. These events indicate that protein synthesis has been restored in the neuronal cell body. In severe damage, although sprouting occurs in axons in the CNS, this process ceases within a short time (about 2 weeks). In this situation, normal functions of the neurons in the CNS are not restored. However, in peripheral nerves, an axon can regenerate satisfactorily if the endoneurial sheaths are intact. In this situation, the regenerating axons reach the correct destination, and the chances of recovery of function are reasonable. The growth rate of an axon has been estimated to be 2 to 4 mm per day.
  • 11. 7-Describe developmental zones of the neural tube during histogenesis of the CNS, describe cell populations originating from the neural crest. The nervous system develops from ectoderm, the surface layer of embryonic tissue. By the third to fourth week of embryonic development, the notochord, of mesodermal origin, induces the development of the neural plate. By the third to fourth week of embryonic development, there is a high rate of cell proliferation. As such, the anterior part of the notochord (of mesodermal origin) begins to thicken, and thus, the neural plate is formed by the third week of fetal life. The neural plate continues to thicken over the following week and expands laterally. As it expands, the faster growing lateral edges of the plate accumulate in a dorsal position as neural folds. As this plate grows and widens, it forms a shallow groove along its longitudinal axis known as the neural groove. The posterior end of the neural plate, which is narrower than the anterior end, will ultimately become the spinal cord, whereas the broader, anterior end will become the brain. As this plate grows and widens, the neural groove becomes deeper. In the process of its forming and deepening, some of the cells located in the lateral margin of the neural groove separate and migrate to a dorsal position to become the neural crest. As the embryo grows, the neural folds fuse along the midline, thus forming a neural tube. The neural tube consists of three layers: 1. an inner layer called the ventricular layer, which is in contact with the cavity of the neural tube; 2. an intermediate layer called the mantle layer; and 3. an outer layer called the marginal layer. The ventricular zone is the major proliferative layer and also the first layer of the forming neural tube to appear. The second layer to form is the marginal layer, followed by the mantle layer. Early in development, the wall of the neural canal becomes thickened, in part, by the formation of young or immature neurons that have yet to completely differentiate (sometimes called neurocytes) in the mantle layer. Because this
  • 12. layer contains the primary cell bodies of neurons, it will ultimately become the gray matter of the spinal cord. Axons associated with cells in the mantle layer will grow into the marginal layer. Histogenesis of CNS: 1. Ventricular zone -separates precursors for neurons and glial cells − -migration of nuclei to base of ventricular cells 2. Marginal zone − -no cell bodies - axons of neurons from intermediate zone invade this zone − 3. Intermediate zone − - forms a interface of ventricular and intermediate zones - no migration of nuclei Cell populations originating from neural crest:  During folding of neural crest, groups of cells appear along neural groove, neural crest cells  Some of these cells give rise sensory ganglia (dorsal root ganglia)  cells of neural crest differentiate into Schwmann cells , pigment cells, meninges and odontoblasts.
  • 13. 8-Describe trophic interactions among the neurons and their target tissue, describe general features of neurotrophic factors. Trophism refers to the ability of certain molecules called trophic (nutritional) factors, to promote cell survival. Neurotrophic factors are polypeptides that support survival, growth, regeneration, and plasticity of neurons. Most types of neuron are generated in excessive numbers, followed later by the death of “surplus” cells soon after axons reach the vicinity of their target. This type of neuronal cell death is regarded to be a consequence of the competition for the limited amount of neurotrophic factors released by target cells (e, g., embryonic muscle cells). This is an adaptive means of adjusting the number of neurons of each type to the number of target cells to be innervated. The “trophic effect” exerted on neurons is illustrated by the trophic influences of “taste nerve fibers” upon the taste buds. Not only do the gustatory nerve fibers convey taste information, but they also have critical roles in both the maintenance and regeneration of taste buds. Following transection of the gustatory nerve fibers, the taste buds degenerate. In time, if and when the transected fibers regenerate into the oral epithelium, new functional taste buds will differentiate from epithelial cells, Presumably, only taste fibers elaborate the essential trophic factors to induce the formation of new taste buds from the oral epithelium. Trophic activity could occur at any time from embryonic life through adulthood. Although a progressive reduction in activity occurs with age, it is never completely lost. In addition to trophic effects, there are tropic effects. Tropism refers to the ability of certain molecules to promote or to guide the outgrowth and directional growth or extension of neuronal processes (axons and dendrites). Neurotrophins are a class among many neurotrophic factors that have important roles in the survival of neurons and have widespread effects throughout the CNS and peripheral nervous system (PNS). Neurotrophins include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin 3 (NT3), and neurotrophin 4/5 (NT 4/5). Examples of related trophic factors include fibroblast growth factor (FGF), the epidermal growth factor family (EGF) and cytokines. The cytokines (e.g., interleukin, a leukemia-inhibitory factor) are extracellular or membrane- anchored polypeptides that mediate communication between cells via cell surface
  • 14. receptors. Trophic factors, as indicated, have roles in promoting the successive stages in the cycle of neuronal differentiation, growth, survival, and programmed cell death 9-Selective neuronal death during ontogenic development of the nervous system; describe a mechanism and its significance. Programme cell death is an integral part of animal tissue development. Development of an organ or tissue is often preceded by the extensive division and differentiation of particular cell, with the resultant mass is then arranged in the correct form with help of apoptosis. Unlike cellular death (caused by hypoxia or other various injury), apoptosis results in cell shrinkage and fragmentation. This allows the cells to be efficiently phagocytosed and ther components removed without releasing of potentially harmful intracellular substances into the surrounding tissue. If a neuron (and especially its DNA) gets damaged by a virus or a toxin, apoptosis destroys and silently removes these sick genes, which may serve to protect surrounding healthy neurons. More importantly, apoptosis is a natural part of development of the immature central nervous system. One of the many wonders of the brain is the built-in redundancy of neurons early in development. These neurons compete vigorously to migrate, innervate target neurons, and drink trophic factors necessary to fuel this process. Apparently, there is survival of the fittest, because up to 50% of many types of neurons normally die in this time of brain maturation. Apoptosis is a natural mechanism to eliminate the unwanted neurons without making as big a molecular mess as doing it via necrosis. Cell death via apoptosis is a prominent feature in mammalian neural development. Recent studies into the basic mechanism of apoptosis have revealed biochemical pathways that control and execute apoptosis in mammalian cells. Protein factors in these pathways play important roles during development in regulating the balance between neuronal life and death. Additionally, mounting evidence indicates such pathways may also be activated during several neurodegenerative diseases, resulting in improper loss of neurons.
  • 15. 10-Describe molecular mechanisms for axon navigation to the target tissue during development and regeneration of the nervous system. Molecular mechanisms of axon guidance: 1. Contact inhibition – adhesion (permissive and non-permissive substrates) 2. Fasciculation (small, involuntary muscle contractions due to spontaneous discharges) 3. Chemotropism (movement caused by chemical stimulus) Cellular and molecular mechanisms for regeneration of nervous system: Different reaction of the CNS and PNS neurons to injury of neural processes. PNS neurons: anabolic processes due to increased synthesis of cytoskeleton and membrane proteins and increased synthesis of RNA. CNS neurons: reaction with distinct regressive mechanisms(atrophy) and destruction of neurons. Decreased synthesis if the RNA
  • 16. 11-Describe structural components of the hematoencephalic barrier, functional significance of HEB.  Meningeal barriers is formed by arachnoid placed between intradural blood vessels and CSF in the subarachnoid space.  Blood-brain barrier, which is located at the interface between the capillary wall and brain tissue. The blood-brain barrier consists of: (1) endothelial cells lining the capillary wall with tight junctions between them, (2) processes of astrocytes abutting on the capillaries as perivascular end-feet, (3) a capillary basement membrane. This arrangement of different cells or their processes prevents the passage of large molecules from the blood into the extracellular space between the neurons and neuroglia and forms the anatomical basis of the blood-brain barrier. One of the beneficial functions of the blood-brain barrier is to prevent entry of blood-borne foreign substances into the brain tissue. However, the existence of this blood-brain barrier also presents a problem when the goal is to deliver drugs into the CNS. In other organs, tight junctions do not exist between the neighboring endothelial cells lining the capillaries.  The capillaries and the ependymal epithelial cells of the choroid plexus form the blood-CSF barrier between the blood and CSF. The presence of this barrier results in distinct differences of concentration of some molecules in the blood and CSF
  • 17. 12-Describe the CNS liquid compartments and their barriers. Fluid (liquid) compartments:  Interstitial fluid: bathing neurons and glial cells within CNS  CSF: in subarachnoid space and ventricular system.  Blood: in the meningeal vessels  Intracellular fluid: in neurons and glial cells CSF: There are four main functions of the CSF. (1) The brain and spinal cord float in the CSF because the specific gravities of these central nervous system (CNS) structures are approximately the same. This buoyant effect of the CSF results in reduction of traction exerted upon the nerves and blood vessels connected with the CNS. (2) The CSF provides a cushioning effect on the CNS and dampens the effects of trauma. (3) The CSF also serves as a vehicle for removal of metabolites from the CNS. (4) Under normal conditions, the CSF provides a stable ionic environment for the CNS. However, the chemical composition of the CSF may change in certain situations such as administration of drugs that cross the blood-brain barrier. The cerebral barriers:  Meningeal barriers: is formed by arachnoid placed between intradural blood vessels and CSF in the subarachnoid space
  • 18. 13- Describe individual mechanisms of transportation throught the hematoencephalic barrier. Transport mechanisms of blood brain barrier (BBB) ⇒ Passive diffusion: lipid soluble substances pass throught the barrier, such as O2, Co2, barbiturates, alcohol ⇒ Facilitated diffusion: substances that penetrate membranes by carrier or mediated mechanisms. This category of transport carries more molecules and rapidly and without consumption of enegy. ⇒ Active transport: consumption of energy, and uses carrier molecules. It proceeds against a concentration gradient. E.g amino acids or extracellular potassium. Moves molecules from the brain and CSF into blood plasma against a concentration gradient. ⇒ Transcytosis: is selective transport by means of the coated vesicles that fuse to form trasnsendothelial canal, e.g. Fe bind transferin ehich has Fe receptors The combination of the specialized cell membrane of the endothelial cells linked by intercellular tight junctions is the hallmark of the BBB. This duo effectively excludes by blocking the passage of many substances across the capillary wall. The permeability property can be enhanced by the state of phosphorylation of the proteins of the cell–cell adherens junctions. The cadherin proteins of the adherens junctions also act as a sig- nalling component between endothelial cells through linkages with the cytoskeletal pro- tein filaments of the endothelial cells. The presence of so few pinocytotic vesicles with- in the endothelial cells is indicative that the transcellular movement by vesicles across the BBB (transendocytosis) is both relatively deficient and slow. However, the selective passage of substances is related to the presence of high concentrations of carriermedi- ated transport systems that act as transporters for glucose, essential amino acids, other required nutrients, and macromolecules. These ensure the passage of essential sub- stances from the blood to the CNSThe combination of the specialized cell membrane of the endothelial cells linked by intercellular tight junctions is the hallmark of the BBB. This duo effectively excludes by blocking the passage of many substances across the ca- pillary wall. The permeability property can be enhanced by the state of phosphorylation of the proteins of the cell–cell adherens junctions. The cadherin proteins of the adherens junctions also act as a signalling component between endothelial cells through linkages with the cytoskeletal protein filaments of the endothelial cells. The presence of so few pinocytotic vesicles within the endothelial cells is indicative that the transcellular move- ment by vesicles across the BBB
  • 19. 14- Describe the CNS structures without total hematoencephalic barrier and their function. There are seven structures in the CNS that lack a blood-brain barrier. Called circumventricular organs, they are the area postrema, pineal body, subcommissural organ, subfornical organ, organum vasculosum of lamina terminalis (OVLT), neurohypophysis (the posterior pituitary gland), and the median eminence. They lack tight junctions in their capillaries. Instead, they have fenestrated capillaries, capillary loops, and large perivascular spaces that permit the passage of larger circulating molecules into the adjacent brain tissue. It is believed that some circulating hormones consisting of large molecules reach their target areas in the brain via the circumventricular organs. For example, the subfornical organ lies in the roof of the third ventricle. Blood-borne angiotensin II reaches the subfornical organ readily because of the lack of the blood-brain barrier in this organ and induces thirst for overall regulation of fluid balance and cardiovascular homeostasis. Functional effects ⇒ Acts as homeostatic organs: e.g neurohypophyses releases oxitocin and vasopressin (ADH) ⇒ Acts as as chemoreceptors e.g. Angiotensin II may act to increase blood pressure ⇒ Acts as osmoreceptors ⇒ The area postrema is the vomiting centre of the brain, detect noxious substances in the blood and stimulate vomiting in order to rid that substances from the body
  • 20. 15- Measuring of the cerebral blood flow The metabolic demands of the brain must be met with the blood supply to this organ. Normal cerebral blood flow is about 50 mL/100 g of brain tissue/min. Thus, a brain of average weight (1500 g) has a normal blood flow of 750 mL/min. Even a brief interruption of the blood supply to the CNS may result in serious neurological disturbances. A blood flow of 25 mL/100 g of brain tissue/min constitutes ischemic penumbra (a dangerously deficient blood supply leading to loss of brain cells). A blood flow of 8 mL/100 g of brain tissue/min leads to an almost complete loss of functional neurons. Consciousness is lost within 10 seconds of the cessation of blood supply to the brain. Freks principle: Cerebral blood flow can be measured by determining the amount of nitrous oxide removed from the blood stream (Qx) per unit of time and dividing that value by the difference between the concentration in the atrial blood (Ax) and the in the venous blood (Vx): Qx CBF= --------------- [Ax] - [Vx] Qx= amount of nitrous oxide removed from the blood Ax= concentration in atrial blood Vx= concentration in the venous blood Average blood flow in young adults is 54ml/100g/min Average brain weight 1400g hence we have a blood flow to brain corresponding to 756ml/min Factors that affect cerebral blood flow : 1) Intracranial pressure 2) Blood viscosity 3) Mean venous pressure 4) Mean atrial pressure 5) Constriction and dilation of cerebral arterioles
  • 21. 16 – Blood flow in various parts of the brain Arterial supply of the brain: Blood supply to the brain is derived from two arteries: (1) the internal carotid artery and (2) the vertebral artery. These arteries and their branches arise in pairs that supply blood to both sides of the brain. The basilar artery is a single artery located in the midline on the ventral side of the brain. The branches of the basilar artery also arise in pairs. Internal Carotid Artery This artery arises from the common carotid artery on each side at the level of the thyroid cartilage and enters the cranial cavity through the carotid canal. Branches: ⇒ The Ophthalmic Artery: enters the orbit through the optic foramen and gives rise to the central artery of the retina, which supplies the retina and cranial dura. Interruption of blood flow in the ophthalmic artery causes loss of vision in the ipsilateral eye. ⇒ The Posterior Communicating Artery: arises at the level of the optic chiasm and travels posteriorly to join the posterior cerebral arteries. Small branches arising from this artery supply blood to the hypophysis, infundibulum, parts of the hypothalamus, thalamus, and hippocampus. ⇒ The Anterior Choroidal Artery: arises near the optic chiasm and supplies the choroid plexus located in the inferior horn of the lateral ventricle, the optic tract, parts of the internal capsule, hippocampal formation, globus pallidus, and lateral portions of the thalamus. ⇒ The Anterior Cerebral Artery: The anterior cerebral artery travels rostrally through the interhemispheric fissure. It supplies blood to the medial aspect of the cerebral hemisphere, including parts of the frontal and parietal lobes. This artery also supplies blood to the postcentral gyrus and precentral gyrus. Occlusion of one of the anterior cerebral arteries results in loss of motor control (paralysis) and loss of sensation in the contralateral leg. Other structures supplied by the anterior cerebral artery include the olfactory bulb and tract, anterior hypothalamus, parts of caudate nucleus, internal capsule, putamen, and septal nuclei. ⇒ The Anterior Communicating Artery: at the level of the optic chiasm, the anterior communicating artery connects the anterior cerebral arteries on the two sides. A group of small arteries arising from the anterior communicating and anterior cerebral arteries penetrates the brain tissue almost perpendicularly and supplies blood to the anterior hypothalamus, including preoptic and suprachiasmatic areas. ⇒ The Medial Striate Artery (Recurrent Artery of Heubner): arises from the anterior cerebral artery at the level of the optic chiasm and supplies blood to the anteromedial part of the head of the caudate nucleus and parts of the internal capsule, putamen, and septal nuclei. The medial striate and the lenticulostriate arteries penetrate the perforated substance. ⇒ The Middle Cerebral Artery: at the level just lateral to the optic chiasm. Branches of the middle cerebral artery supply blood to the lateral convexity of the cerebral hemisphere including parts of the temporal, frontal, parietal, and occipital lobes.
  • 22. Vertebro-Basilar Circulation: This system includes the two vertebral arteries, the basilar artery (which is formed by the union of the two vertebral arteries), and their branches. This arterial system supplies the medulla, pons, mesencephalon, and cerebellum. Braches: ⇒ The vertebral artery: on each side is the first branch arising from the subclavian artery. It enters the transverse foramen of the sixth cervical vertebrae, ascends through these foramina in higher vertebra, and eventually enters the cranium through the foramen magnum. In the cranium, at the medullary level, each vertebral artery gives off the anterior spinal artery, the posterior inferior cerebellar artery, and the posterior spinal artery. ⇒ The Anterior Spinal Artery: at the confluence of the two vertebral arteries, two small branches arise and join to form a single anterior spinal artery. This artery supplies the medial structures of the medulla, which include the pyramids, medial lemniscus, medial longitudinal fasciculus, hypoglossal nucleus, and the inferior olivary nucleus. ⇒ The Posterior Inferior Cerebellar Artery (PICA): arises from the vertebral artery and supplies the regions of the lateral medulla that include the spinothalamic tract, dorsal and ventral spinocerebellar tracts, descending sympathetic tract, descending tract of cranial nerve V, and nucleus ambiguus. ⇒ The Posterior Spinal Artery (PSA): It is the first branch of the vertebral artery in the cranium in about 25% of cases. However, in a majority of cases (75%), it arises from the posterior inferior cerebellar artery. In the caudal medulla, this artery supplies the fasciculus gracilis and cuneatus as well as the gracile and cuneate nuclei, spinal trigeminal nucleus, dorsal and caudal portions of the inferior cerebellar peduncle, and portions of the solitary tract and dorsal motor nucleus of the vagus nerve. ⇒ The Basilar Artery: The two vertebral arteries join at the caudal border of the pons to form the single basilar artery. ⇒ The Anterior Inferior Cerebellar Artery (AICA): is the most caudal branch arising from the basilar artery. The AICA supplies the ventral and inferior surface of the cerebellum and lateral parts of the pons. ⇒ The labyrinthine (internal auditory) artery: is usually a branch of the AICA and supplies the cochlea and labyrinth. ⇒ The Pontine Arteries: Several pairs of pontine arteries arise from the basilar artery. Some pontine arteries (the paramedian arteries) enter the pons immediately and supply the medial portion of the lower and upper pons. Some pontine arteries (the short circumferential arteries) travel a short distance around the pons and supply substantia nigra and lateral portions of the midbrain tegmentum. ⇒ The superior cerebellar artery: arises just caudal to the bifurcation of the basilar artery and supplies the rostral level of the pons, caudal part of the midbrain, and superior surface of the cerebellum. ⇒ The posterior cerebral arteries arise at the terminal bifurcation of the basilar artery. Branches of the posterior cerebral arteries supply most of the midbrain, thalamus, and subthalamic nucleus. Cerebral Arterial Circle (Circle of Willis)
  • 23. The cerebral arterial circle surrounds the optic chiasm and the infundibulum of the pituitary. It is formed by the anastomosis of the branches of the internal carotid artery and the terminal branches of the basilar artery. The anterior communicating artery connects the two anterior cerebral arteries, thus forming a semicircle. The circle is completed as the posterior communicating arteries arising from the internal carotid arteries at the level of the optic chiasm travel posteriorly to join the posterior cerebral arteries that are formed by the bifurcation of the basilar artery. The circle of Willis is patent in only 20% of individuals. When it is patent, this arterial system supplies the hypothalamus, hypophysis, infundibulum, thalamus, caudate nucleus, putamen, internal capsule, globus pallidus, choroid plexus (lateral ventricles), and temporal lobe.
  • 24. 17- Regulation of cerebral circulation. Brain metabolism. Brain metabolism: 3 metabolic factors have potent effect on control of cerebral blood flow (CBF) : ⇒ Increase in [CO2] leads to increase of CBF. CO2 + H2O  H2CO3  HCO3- + H+ AND the H+ causes the dilation of cerebral vessels ⇒ Increase in H+ leads to increase in CBF ⇒ Decrease in O2 leads increase CBF via vasodilation The brain (2% of the total body weight) receives about 15% of the cardiac output and consumes about 20% of the total O2 consumption. The brain is highly sensitive to disturbances of the blood supply. Ischaemia lasting seconds causes symptoms and lasting for few minutes causes irreversible damage. The caliber of the arterioles is regulated by: ⇒ Local vasodilators; metabolites (e.g. CO2)~ ⇒ Vasoactive substances produced by the endothelium, circulating peptides such as angiotensin II ⇒ Vasomotor nerves ⇒ Autoregulation mechanisms (the systemic blood pressure increases, but the cerebral blood flow remains constant by: a) intraluminar pressure within the arterioles elicits direct myogenic responde b) hypocapnia causes arterial vasoconstriction Intracranial pressure: ⇒ cerebral vessels are compressed even if there is no increase in intracranial pressure ⇒ any change in venous pressure causes a similar change in intracranial pressure. A rise in venous pressure decreases cerebral blood flow. ⇒ Cerebreal circulation has strong sympathetic innervation extending from the superior cervical ganglion ⇒ Brain extremely sensitive to hypoxia ⇒ Ammonia very toxic to nerve cells and leaves the brain in the form of glutamine
  • 25. 18- Formation and absorption of cerebrospinal fluid. Function of cerebrospinal fluid. Formation of the Cerebrospinal fluid: About 70% of the CSF present in the brain and spinal cord is produced by the choroid plexuses. The remaining 30% of CSF, which is secreted by the parenchyma of the brain, crosses the ependyma (a single layer of ciliated columnar epithelial cells lining the ventricular system) and enters the ventricles. The formation of CSF is an active process involving the enzyme carbonic anhydrase and specific transport mechanisms. The formation of the CSF first involves filtration of the blood through the fenestrations of the endothelial cells that line the choroidal capillaries. However, the movement of peptides, proteins, and other larger molecules from this filtrate into the CSF is prevented by the tight junctions that exist in the neighboring epithelial cells that form the outer layer of the choroid plexus. Energy-dependent active transport mechanisms are present in the choroidal epithelium for transporting Na+ and Mg2+ ions into the CSF and for removing K+ and Ca2+ ions from the CSF. Water flows across the epithelium for maintaining the osmotic balance. Normally, the rate of formation of CSF is about 500 mL/day and the total volume of CSF is 90 to 140 mL, of which about 23 mL is in the ventricles, and the remaining is in the subarachnoid space. Circulation: The movement of CSF is pulsatile. It flows from the lateral ventricles into the third ventricle through the foramina of Monro where it mixes with more CSF. Then, it flows through the cerebral aqueduct (aqueduct of Sylvius) into the fourth ventricle, where additional CSF is secreted. The fluid leaves the ventricular system via the foramina of Luschka and Magendie and enters the cerebellomedullary cistern (cisterna magna). The CSF then travels rostrally over the cerebral hemisphere where it enters the arachnoid villi. Absorption: is made throught the arachidonic vili. The CSF drains into dural venous sinuses, there are valves here, so fluid flows only from vili to veins where pressure difference is appropriate. Functions: There are four main functions of the CSF. (1) The brain and spinal cord float in the CSF because the specific gravities of these central nervous system (CNS) structures are approximately the same. This buoyant effect of the CSF results in reduction of traction exerted upon the nerves and blood vessels connected with the CNS. (2) The CSF provides a cushioning effect on the CNS and dampens the effects of trauma. (3) The CSF also serves as a vehicle for removal of metabolites from the CNS. (4) Under normal conditions, the CSF provides a stable ionic environment for the CNS. Composition: Normally, very little protein is present in the CSF, and this is the primary difference between CSF and blood serum. The concentrations of glucose, as well as
  • 26. 19 – Resting potential of the neuron Resting membrane potential: When a neuron is not generating action potentials, it is at rest. When the neuron is at rest, its cytosol along the inner surface of its membrane is negatively charged compared with the charge on the outside. Typically, the resting membrane potential (or resting potential) of a neuron is -65 millivolts (mV). The potential difference across the cell membrane during resting state is called the resting membrane potential. The lipid bilayer of the neuronal membrane maintains this separation of charges by acting as a barrier to the diffusion of ions across the membrane. The ion concentration gradients across the neuronal membrane are established by ion pumps that actively move ions into or out of neurons against their concentration gradients. The selective permeability of membranes is due to the presence of ion channels that allow some ions to cross the membrane in the direction of their concentration gradients. The ion pumps and ion channels work against each other in this manner. If the neuronal membrane is selectively permeable to only a K+ ion, this ion will move out of the neuron down its concentration gradient. Therefore, more positive charges accumulate outside the neuron. The fixed negative charges inside the neuron impede the efflux of positively charged K+ ions, and excess positive charges outside the neuron tend to promote influx of the K+ ions into the neuron due to the electrostatic forces. The opposite charges attract, while similar charges repel each other. Thus, two forces are acting on the flow of K+ ions out of the neuron; a higher concentration inside the neuron (concentration gradient) tends to expel them out of the neuron, while the electrostatic forces tend to prevent their flow out of the neuron. When the two opposing forces are equal, K+ concentrations inside and outside the neuron are in equilibrium. The value of the membrane potential at this time is called the K+ equilibrium potential. Thus, if the neuronal membrane contained only K+ channels, the resting membrane potential would be determined by the K+ concentration gradient and would be equal to the equilibrium potential for K+ ions (approximately -80 mV). However, as stated earlier, the resting membrane potential of a neuron is usually -65 mV. This is because neurons at rest are permeable to the Na+ ion also. The Na+ ions tend to flow into the neuron due to two forces: (1) concentration gradient of Na+ ions (extracellular Na+ concentration is much higher than its intracellular concentration) and (2) electrostatic forces (there is an excess of positive charges outside and an excess of negative charges inside the neuron). Due to the influx of Na+ ions, the resting membrane potential deviates from that of the K+ equilibrium potential (i.e., it becomes -65 mV instead of -80 mV). However, the membrane potential does not reach the equilibrium potential for + Na . The reason for the neuron's inability to attain a resting membrane potential closer to the Na+ equilibrium potential is that the number of open nongated Na+ channels is much smaller than the number of open nongated K+ channels in the resting state of a neuron. The permeability of Na+ is small despite large electrostatic and concentration gradient forces tending to drive it into the neuron. To maintain a steady resting membrane potential, the separation of charges across the neuronal membrane must be maintained at a constant. This is accomplished by the Na+-K+ pump described earlier. Goldman equation: since the neuronal membrane is permeable to more than one ion, the goldman equation is used to calculate membrane potential. This equation takes into account the contribution of the permeability of each ion and its extra- and intracellular concentration. Nerst equation: is used to calculate equilibrium potential of an ion that is present on both sides of the cell membrane.
  • 27. 20- Receptor, synaptic and action potential-description Receptor potential: whatever the stimulus that excites the receptor, its immediate effect is to arrange the membrane potential of the receptor. This change is called receptor potential. Different receptors can be excited, either: a) By mechanical deformation that stretches the receptor membrane and opens ion channels. b) By application of a chemical to the membrane c) By change of temperature d) By eletromagnetic radiation When the receptor potential rises above the threshold for eliciting an action potential, the the action potential begins to appear. The more the receptor potential rises above the threshold level, the greater becomes the action potential frequency Synaptic potential: an interaction of a transmitter on postsynaptic neuron initiates a synaptic potential. Can be either: EPSP (excitatory postsynaptic potential) IPSP (inhibitory postsynaptic potential) Action potential: rapid change in the membrane potential. It begins with a sudden change from the normal resting membrane potential to a positive membrane potential. When a neuron receives an excitatory input, the neuronal membrane is depolarized, resulting in an opening of some voltage-gated Na+ channels and influx of Na+ . The accumulation of positive charges due to influx of Na+ promotes depolarization of the neuronal membrane. When the membrane potential reaches threshold potential, the chances of generating an action potential are about 50%. However, when the membrane is depolarized beyond the threshold potential, a sufficient number of voltage- gated Na+ channels open, relative permeability of Na+ ions is greater than that of K+ ions, and action potentials are generated with certainty. During the rising phase of the action potential, there is a rapid depolarization of the membrane due to increased permeability of Na+. The depolarization continues so that the membrane potential approaches the Na+ equilibrium potential. The part of the action potential where the inside of the neuron is positive relative to the outside is called the overshoot. Towards the end of the rising phase of the action potential, voltage-gated Na+ channels are inactivated, and the influx of Na+ through these channels is stopped. During the falling phase of the action potential, the neuron is repolarized by opening of voltage-gated K+ channels, which allows increased efflux of K+ from the neuron through these channels. The opening of voltage-gated K+ channels is also caused by depolarization of the neuronal membrane. Because these voltage-gated K+ channels open with a delay (about 1 msec) after the membrane depolarization and their opening rectifies the membrane potential, they are called delayed rectifier K+ channels. At the end of the falling phase, the membrane potential is more negative than the resting potential because of increased K+ permeability caused by the opening of the delayed rectifier K+ channels in addition to the already present resting K+ permeability through nongated channels. The permeability is closer to the equilibrium potential of K+ because there is little Na+ permeability during this period. This portion of the action potential is called after-hyperpolarization or undershoot. Once after-hyperpolarization has occurred, the resting membrane potential is restored gradually as the voltage-gated K+ channels close again.
  • 28. 21 – Ionic basis of membrane potential changes The resting membrane potential of a neuron is usually -65 mV. At rest, Na+ influx into the neuron through open nongated Na+ channels is balanced by the efflux of K+ through open nongated K+ channels. Thus, the membrane potential remains constant closer (but not equal) to the K+ equilibrium. When a neuron receives an excitatory input, the neuronal membrane is depolarized, resulting in an opening of some voltage-gated Na+ channels and influx of Na+. Na+ channels are normally closed. The accumulation of positive charges due to influx of Na+ promotes depolarization of the neuronal membrane. When the membrane potential reaches threshold potential, the chances of generating an action potential are about 50%. However, when the membrane is depolarized beyond the threshold potential, a sufficient number of voltage-gated Na+ channels open, relative permeability of Na+ ions is greater than that of K+ ions, and action potentials are generated with certainty. Generation of an action potential is an all-or-nothing phenomenon. Because the concentration of Na+ channels is relatively high at the axon hillock, this is the site of generation of action potentials in a neuron. During the rising phase of the action potential generation, there is a rapid depolarization of the membrane due to increased permeability of Na+. The depolarization continues so that the membrane potential approaches the Na+ equilibrium potential. The part of the action potential where the inside of the neuron is positive relative to the outside is called the overshoot. Towards the end of the rising phase of the action potential, voltage-gated Na+ channels are inactivated, and the influx of Na+ through these channels is stopped. During the falling phase of the action potential, the neuron is repolarized by opening of voltage-gated K+ channels, which allows increased efflux of K+ from the neuron through these channels. The opening of voltage-gated K+ channels is also caused by depolarization of the neuronal membrane. Because these voltage-gated K+ channels open with a delay (about 1 msec) after the membrane depolarization and their opening rectifies the membrane potential, they are called delayed rectifier K+ channels. At the end of the falling phase, the membrane potential is more negative than the resting potential because of increased K+ permeability caused by the opening of the delayed rectifier K+ channels in addition to the already present resting K+ permeability through nongated channels. The permeability is closer to the equilibrium potential of K+ because there is little Na+ permeability during this period. This portion of the action potential is called after-hyperpolarization or undershoot. Once after-hyperpolarization has occurred, the resting membrane potential is restored gradually as the voltage-gated K+ channels close again. The sodium channel exists in the following three states: resting, activated, or inactivated. • Resting state: During this state, the activation gate closes the channel pore while the inactivation gate is open. With the channel pore closed, Na+ cannot flow into the neuron. • Activated state: During the rising phase of action potential, both activation and inactivation gates are open, and Na+ ions flow into the neuron. • Inactivated state: During this state, the inactivation gate closes the channel pore while the activation gate is still open. Even though the activation gate is open, Na+ cannot flow into the neuron. The neuron cannot be activated until the
  • 29. 22- Ion channels in neurons – their distributions Ion Channels: . Ion channels are made up of proteins that are embedded in the lipid bilayer of the neuronal membrane across which they span. They are characterized by the following general properties. • The flow of ions through the channels does not require metabolic energy; the flow is passive. • The electrochemical driving force across the membrane, but not the channel itself, determines the direction and eventual equilibrium of this flow. • The ionic charge determines whether a channel allows an ion to flow through; some channels allow cations while others allow anions to flow through them. • Most cation-selective channels allow only one ion species (e.g., Na+ or K+ or Ca2+) to flow through them. However, some channels allow more than one ion species to flow through them. For example, when L-glutamate (an excitatory amino acid neurotransmitter) activates an N-methyl-D-aspartic acid (NMDA) receptor, both Na+ and Ca2+ ions flow through the NMDA receptor channel into the neuron. • Most anion-selective channels allow only Cl- to flow through them. • Some blockers can prevent the flow of ions through the ion channels. For example, phencyclidine (PCP, or Angel Dust) blocks the NMDA receptor channel. Classification of Ion Channels: Nongated Channels: Although nongated channels are capable of opening as well as closing, most of the time they are in the open site. They control the flow of ions during the resting membrane potential. Examples include nongated Na+ and K+ channels that contribute to the resting membrane potential. Gated Channels: These channels are also capable of opening as well as closing. All gated channels are allosteric proteins The channels that are opened or closed by a change in the membrane potential are called voltage-gated channels. The opening and closing of the channel is believed to be due to the movement of the charged region of the channel back and forth through the electrical field of the membrane. Voltage-gated channels exist in three states: (1) resting state (the channel is closed but can be activated) (2) active state (the channel is open), and (3) refractory state (the channel is inactivated). Changes in the electrical potential difference across the membrane provide the energy for gating in these channels. Genes encoding for voltage-gated Na+, K+, and Ca2+ channels belong to one family. These channels are described as follows. The voltage-gated Na+ channel is formed by a single long polypeptide (a string of amino acids containing peptide bonds) that has four domains (I-IV). Each domain has six hydrophobic alpha helices (S1aS6) that span back and forth within the cell membrane. The four domains join together and form an aqueous pore of the channel. An additional hydrophobic region connects the S5 and S6 alpha helical segments,
  • 30. forming a pore loop. The presence of this pore loop makes the channel more permeable to Na+ than to K+. The membrane-spanning S4 alpha helical segment is believed to be voltage sensitive. At the resting membrane potential, the channel pore is closed. The S4 segment undergoes a conformational change when the membrane potential changes (e.g., when the neuron is depolarized), the S4 segment is pushed away from the inner side of the membrane, and the channel gate opens, allowing an influx of Na + ions. There are some cases where Na+ permeability is blocked. Tetrodotoxin (TTX), a toxin isolated from the ovaries of Japanese puffer fish, binds to the sodium channel on the outside and blocks the sodium permeability pore. Consequently, neurons are not able to generate action potentials after the application of TTX. These channels are also blocked by local anesthetic drugs (e.g., lidocaine). The basic structure of the voltage-gated Ca2+ channel is similar to that of the voltage-gated Na+ channel. Ca2+ ions enter the postsynaptic neurons through these channels and activate enzymes. Depolarization of presynaptic nerve terminals results in entry of Ca2+ ions into the terminal via these channels. An increase in the levels of intracellular Ca2+ results in the release of transmitters from presynaptic nerve terminals. Different varieties of voltage-gated K+ channels have been identified, and they serve different functions. The general scheme describing the components of this channel is similar to that of the voltage-gated Na+ channel, except that the voltage-gated K+ channel consists of four polypeptides. It should be recalled that each polypeptide contributing to the formation of a large protein molecule is called a subunit. Each subunit of a voltage-gated K+ channel consists of six alpha-helical membrane-spanning segments (S1 a S6). A pore loop makes the channel more permeable to K + than to Na+. The S4 segment acts as an activation gate. The K+ channels are generally blocked by chemicals such as tetraethylammonium (TEA) or 4-aminopyridine. The ligand-gated channels are opened by noncovalent binding of chemical substances with their receptors on the neuronal membrane. These chemical substances include: (1) transmitters or hormones present in the extracellular fluid that bind to their receptors on the extracellular side of the channel and bring about a conformational change to open the channel (e.g., acetylcholine, γ-aminobutyric acid [GABA], or glycine); and (2) an intracellular second messenger (e.g., cyclic adenosine monophosphate, which is activated by a transmitter such as norepinephrine). The second messenger can open the channel (1) directly by binding to the channel and causing a conformational change or (2) indirectly by phosphorylating the channel protein in the presence of a protein kinase and causing a conformational change; this effect on the channel is reversed by dephosphorylation catalyzed by a protein phosphatase. Genes encoding for transmitter-gated channels (e.g., channels activated by acetylcholine, GABA, or glycine) and genes encoding for voltage-gated channels belong to different families. Mechanically gated channels open by a mechanical stimulus and include the channels involved in producing generator potentials of stretch and touch receptors. 23 – Spreading of membrane potentials. Length and time constant of the membrane. An action potential elicited at any one point on an excitable membrane usually excites adjacent portions of the membrane, resulting in the propagation of the action potential.
  • 31. A nerve fiber excited at its midportion-develop increased permeability to Na+. Positive electrical charges carried by the inward diffusing Na+ flow inside the fiber throught depolarized membrane and then for several milimiters along the core of the axon. These positive cgarges increase the voltage to above the thereshold. Thus the depolarization process travels along the entire extent of the fiber and the transmission of this depolarization process is called nerve impulse. The action potential will travel in both directions away from the stimulus until the entire membrane becomes depolarized. Once an action potential has been elicited at any point on the membtrane of a normal fiber, the depolarization process travels over the entire membrane if conditions are right, or it does not travel at all if conditions are not right- this is called “all or nothing principle”, and it applies to all normal excitable tissues. Occasionally, the action potential reaches a point on the membrane at which it does not generate voltage to stimulate the next area of the membrane. When this occurs, the spread of depolarization stops. Therefore, for continued propagation of an impulse , the ratio of action potential to threshold for excitation must at all times be greater than 1 – safety factor for propagation. Length-constant: measures the effectiveness of neuron in longitudinal signal transduction. Rm λ= √ ------------------- (Ri + Ro ) SOFIA: lambda e igual a raiz quadrada de abrir parentesis Rm sobre abrir parentesis ( Ri + Ro ) Rm: is the resistance across the membrane Ri: resistance inside the membrane Ro: is the resistance outside the membrane The larger the length constant the bigger the effect of the action potential Long leght constant results in spatial summation Time constant: T= Rm . Cm R: resistance C: capacitance Long-time constant results in temporal summation 24 – Temporal summation of membrane potentials. Temporal summation: is an effect generated by a single neuron as a way of achieving action potential. Summation occurs when the time constant is sufficiently long and the frequency of rises in potential are high enough that a rise in potential
  • 32. begins before a previous one ends. The amplitude of the previous potential at the point where the second begins will algebraically summate, generating a potential that is overall lager than the individual potentials. This allows the potential to reach the thereshold to generate the action potential. Thus successive postsynaptic potentials caused by discharges from a single pre- synaptic, if they occur rapidly enough, can summate in the same way that postsynaptic potentials may summate from widely distributed terminals over the surface of a neuron. The degree of temporal summation is directly proportional to time – constant; and it occurs when the second potential arrises before the 1st has decreased. Temporal summation is involved in vision. The inverse proportion of intensity and time, applies as long as the stimulus is no greater than 0.1 second. For example, at 0.1 second, 130 quanta are absorbed, un any matter of provision, but when raised to 1 second there is a lesser rate of summation, needing 230 quanta to compensate for the decrease in intensity. The frequency of vision is function of frequency of flashes, so the longer the stimulus, the better chance it can attain the number of quanta needed for vision. 25 – Conduction velocity of the action potential, its determiants. When a region of an unmyelinated axonal membrane is depolarized sufficiently by a depolarizing stimulus (e.g., a synaptic potential in a neuron) to reach a threshold
  • 33. potential, voltage-gated Na+ channels open, Na+ flows into the axoplasm, and an action potential is generated in that region of the axon. Some of the current generated by the action potential spreads by electrotonic conduction (passive spread) to an adjacent region of the axon. The passive spread of current occurs by movement of electrons, and movement of Na+ ions is not required. At the adjacent region, the passive spread of current results in opening of voltage-gated Na+ channels, influx of Na+ into the axoplasm, and generation of an action potential. In other words, the passive spread of voltage along the length of an axon results in an active regeneration process. The propagation of an action potential along the axon depends on the cable properties of the axon. The larger the diameter of the axon, the lower the resistance there is to the flow of current along its length. Therefore, the conduction velocity (propagation of action potential) along the length of the axon can be increased by increasing its diameter. For example, the axons of stellate ganglion neurons in the squid are about 1 mm in diameter (1000 times larger than the axons of mammalian neurons). The conduction of action potential in these squid giant axons is faster than in mammalian axons. The squid needs these fast conducting axons for faster contraction of the mantle muscles that produce a jet propulsion effect needed for quick escape from predators. In vertebrates, the conduction velocity is increased by myelination of axon. A myelin sheath consists of about 1-mm lengths of as many as 300 concentric layers of membrane around a single axon. In the peripheral nervous system, myelin is formed by Schwann cells. In the central nervous system, oligodendrocytes form the myelin. Nodes of Ranvier (bare segments of the axonal membrane with a very high density of voltage- gated Na+ channels) are present in between the segments of the myelin sheath. The myelinated segments of an axon are not excitable and have a high resistance to the leakage of current across them. On the other hand, passive spread of current can generate an intense current at the nodes of Ranvier due to the presence of a high density of voltage-gated Na+ channels. When a depolarizing stimulus (e.g., a synaptic potential in a neuron) arrives at a node of Ranvier, Na+ channels open, there is an influx of Na+ ions, and an action potential is generated at that node. Some current generated by the action potential spreads passively to the next node of Ranvier, and depolarization of the membrane at this node results in the generation of an action potential. By this time, Na+ channels at the preceding node are inactivated, K+ channels open, and repolarization occurs. Thus, the action potential propagates along a myelinated axon by saltatory conduction (i.e., the jumping of an action potential from one node to another). Myelination of an axon has two advantages: (1) conduction is very rapid along an axon, and (2) there is a conservation of metabolic energy because excitation is restricted to the nodal regions that are relatively small (0.5 µm). Conduction is also influenced by temperature, a high temperature leads to a higher conduction velocity We can also note that the spinocerebellar tract has highest conduction velocity. 26 – Electrical and chemical transmission at synapses. Types of Synaptic Transmission:
  • 34. Two types of synaptic transmission electrical and chemical are recognized in the nervous system. It should be noted that the electrical synapses are relatively less common than the chemical synapses in the mammalian nervous system. Electrical Transmission In electrical transmission between the nerve cells, the current generated by an impulse in one neuron spreads to another neuron through a pathway of low electrical resistance. Electrical synapses occur at gap junctions. In an electrical synapse, ion channels connect the cytoplasm of the presynaptic and postsynaptic cells. In the adult mammalian central nervous system, electrical synapses are present where the activity of neighboring neurons needs to be highly synchronized. For example, hormone-secreting neurons in mammalian hypothalamus are connected with electrical synapses so that they fire almost simultaneously and secrete a burst of hormone into the circulation. At an electrical synapse, the current generated by voltage-gated channels at the presynaptic neuron flows directly into the postsynaptic neuron. Therefore, transmission at such a synapse is very rapid (<0.1 msec). At some synapses (e.g., in the giant motor synapse of crayfish), the current can pass in one direction (from presynaptic to postsynaptic neuron) but not in the reverse direction. Such synapses are called rectifying or unidirectional synapses. At other synapses, the current can pass equally well in both directions. Such synapses are called nonrectifying or bidirectional synapses. Most electrical synapses in mammalian nervous system are believed to be the nonrectifying type. Chemical Transmission: At chemical synapses, there is no continuity between the cytoplasm of the presynaptic terminal and postsynaptic neuron. Instead, the cells are separated by synaptic clefts, which are fluid-filled gaps (20-50 nm). The presynaptic and postsynaptic membranes adhere to each other due to the presence of a matrix of extracellular fibrous protein in the synaptic cleft. The presynaptic terminal contains synaptic vesicles that are filled with several thousand molecules of a specific chemical substance, the neurotransmitter. Pyramid-like structures consisting of proteins arise from the intracellular side of the presynaptic terminal membrane and project into the cytoplasm of the presynaptic terminal. These pyramids and the membranes associated with them are called active zones and are the specialized release sites in the presynaptic terminal. The vesicles containing the neurotransmitter are aggregated near the active zones. Mechanisms of Transmitter Release: An action potential depolarizes the presynaptic nerve terminal, voltage-gated Ca channels located in the presynaptic terminal membrane open, Ca2+ permeability 2+ increases, and Ca2+ enters the terminal. These events cause the membrane of the 27 – Excitatory and inhibitory neurotrasmitters. Neurotrasmitter: chemical substance that is synthesized in a neuron, released at a synapse following depolarization of the nerve terminal (usually dependent on influx of
  • 35. calcium ions), which binds to receptors on the postsynaptic cell and/or presynaptic terminal to elicit a specific response. (1) the substance must be synthesized in the neuron, and the enzymes needed for its synthesis must be present in the neuron; Small Molecule it must be released in sufficient quantity to elicit a response from the (2) Gaseous Neurotransmittersneuron or cell located in the effector Neurotransmitters postsynaptic Neuropeptides organ; Acetylcholine(3) mechanisms forOpioid peptides Nitric oxide neurotransmitter from the removal or inactivation of the Excitatory amino acids exist; and β-endorphin, synaptic cleft must Glutamate (4) it should mimic the action of the endogenously released neurotransmitter Methionine- Aspartate administered exogenously at or near a synapse. when enkephalin Inhibitory amino acids Leucine-enkephalin GABA Endomorphins Glycine Nociceptin Classes of Biogenic amines Substance P Neurotrans- Catecholamines mitters Dopamine Norepinephrine Epinephrine Indoleamine In the CNS, a neuron is contantly bombarded by neurotrasmitters, each of which Serotonin (5- can generate or modify a synaptic potential. hydroxytryptamine, [5-HT]) Neurotrasmitters that move the membrane potential towars depolarization with Imidazole amine the resultant production of an action potential are known as excitatory neurotrasmitters. Histamine Neurotrasmitters that move the membrane away from depolarization by making Purines the resting membrane potential more negative, the membrane is hyperrepolariyed, are ATP known as inhibitory neurotrasmitters. Adenosine Because the postsynaptic response is actually elicited by the receptor rather than by the trasmitters, the postsynaptic receptor determines whether a given neurotransmitter will be excitatory or inhibitory. Some neurotrasmitters can have either effect, depending on the type of postsynaptic receptor present. Excitatory: Acetylcholine, glutamate, aspratate.. Inhibitory: Dopamine, adenosine, serotonin, histamine, GABA… Both: Epinepherine, norepinepherine, glycine Acetylcholine Neurotransmitter in both the peripheral nervous system (PNS) and central nervous system (CNS). Acetylcholine is one of many neurotransmitters in the autonomic nervous system (ANS) and the only neurotransmitter used in the somatic nervous system. It is also the neurotransmitter in all autonomic ganglia. In the peripheral nervous system, acetylcholine activates muscles, and is a major neurotransmitter in the autonomic nervous system. When acetylcholine binds to acetylcholine receptors on skeletal muscle fibers, it opens ligand gated sodium channels in the cell membrane. Sodium ions then enter the muscle cell, stimulating muscle contraction. Acetylcholine, while inducing contraction of skeletal muscles, instead
  • 36. induces decreased contraction in cardiac muscle fibers. This distinction is attributed to differences in receptor structure between skeletal and cardiac fibers. In the autonomic nervous system, acetylcholine is released in the following sites: • all pre- and post-ganglionic parasympathetic neurons • all preganglionic sympathetic neurons o preganglionic sympathetic fibers to suprarenal medulla, the modified sympathetic ganglion; on stimulation by acetylcholine, the suprarenal medulla releases epinephrine and norepinephrine • some postganglionic sympathetic fibers o sudomotor neurons to sweat glands. In the central nervous system, ACh has a variety of effects as a neuromodulator, e.g., for plasticity and excitability. Other effects are arousal and reward. Damage to the cholinergic system in the brain has been suggested to play a role in the memory deficits associated with Alzheimer's Disease. Types of acetylcholine receptors: • Nicotinic AChRs are ionotropic receptors permeable to sodium, potassium, and chloride ions. They are stimulated by nicotine and acetylcholine. They are of two main types, muscle type and neuronal type. The former can be selectively blocked by curare and the latter by hexamethonium. The main location of nicotinic AChRs is on muscle end plates, autonomic ganglia (both sympathetic and parasympathetic), and in the CNS. • Muscarinic receptors are metabotropic, and affect neurons over a longer time frame. They are stimulated by muscarine and acetylcholine, and blocked by atropine. Muscarinic receptors are found in both the central nervous system and the peripheral nervous system, in heart, lungs, upper GI tract and sweat glands. Glutamate: Some of the important physiological and clinical considerations relevant to glutamate are as follows. • Glutamate has been implicated as a transmitter in a variety of circuits in the brain. E.g. excitatory amino acids may be involved in learning and memory processes, as well as motor functions. Dopamine is a neurotransmitter occurring in a wide variety of animals. In the brain, it functions as a neurotransmitter, activating the five types of dopamine receptors
  • 37. — D1, D2, D3, D4 and D5, and their variants. Dopamine is produced in several areas of the brain, including the substantia nigra and the ventral tegmental area. Dopamine is also a neurohormone released by the hypothalamus. Its main function as a hormone is to inhibit the release of prolactin from the anterior lobe of the pituitary. Epinepherine: when in the bloodstream, it rapidly prepares the body for action in emergency situations. The hormone boosts the supply of oxygen and glucose to the brain and muscles, while suppressing other non-emergency bodily processes. It increases heart rate and stroke volume, dilates the pupils, and constricts arterioles in the skin and gastrointestinal tract while dilating arterioles in skeletal muscles. It elevates the blood sugar level by increasing catabolism of glycogen to glucose in the liver, and at the same time begins the breakdown of lipids in fat cells. Like some other stress hormones, epinephrine has a suppressive effect on the immune system. Epinephrine's actions are mediated through adrenergic receptors. Epinephrine is a non-selective agonist of all adrenergic receptors. It activates α1, α2, β1, and β2 receptors to different extents. Specific functions include: • It binds to α1 receptors of liver cells, which activate inositol-phospholipid signaling pathway, signaling the phosphorylation of glycogen synthase and phosphorylase kinase (inactivating and activating them, respectively), leading to the latter activating another enzyme—glycogen phosphorylase—which catalises breakdown of glycogen (glycogenolysis) so as to release glucose to the bloodstream. Simultaneously protein phosphatase-1 (PP1) is inactivated, as in the active state PP1 would reverse all the previous phosphorylations. • Epinephrine also activates β-adrenergic receptors of the liver and muscle cells, thereby activating the adenylate cyclase signaling pathway, which will in turn increase glycogenolysis. β2 receptors are found primarily in skeletal muscle blood vessels where they trigger vasodilation. However, α-adrenergic receptors are found in most smooth muscles and splanchnic vessels, and epinephrine triggers vasoconstriction in those vessels. Epinepherine is found in the spinal cord, hypothalamus, thalamus and periaqueducts. Serotonin: Serotonin-containing cells in the raphe regions of the brainstem are believed to play a role in descending pain-control systems. Other serotonin-containing neurons may play a role in mediating affective processes such as aggressive behavior and arousal. Serotonin synthesized in the pineal gland serves as a precursor for the synthesis of melatonin, which is a neurohormone involved in regulating sleep patterns. Serotonin is also believed to play an important role in depression. Histamin: Histamine has been implicated as a transmitter in the regulation of food and water intake, as well as in thermoregulation and autonomic functions.
  • 38. GABA: • GABA is an inhibitory transmitter in many brain circuits. E.g, GABA is used as an inhibitory neurotransmitter by the Purkinje cells in the cerebellum. Alteration of GABAergic circuits has been implicated in neurological and psychiatric disorders like Huntington's chorea, Parkinson's disease, senile dementi… • Therefore, dietary deficiency of vitamin B6 can lead to diminished GABA synthesis. In a disastrous series of infant deaths.. GABA content in the brain of these infants was reduced. Subsequently, there was a loss of synaptic inhibition that caused seizures and death. • Since epileptic seizures can be facilitated by lack of neuronal inhibition, increase in the inhibitory transmitter, GABA, is helpful in terminating them • Barbiturates act as agonists or modulators on postsynaptic GABA receptors and are used to treat epilepsy. 28 – Excitatory and inhibitory postsynaptic potentials. Postsynaptic potentials are changes in the membrane potential of the postsynaptic terminal of a chemical synapse. Postsynaptic potentials are graded potentials, and their function is to initiate or inhibit action potentials. They are caused by the presynaptic neuron releasing neurotransmitters from the terminal button at the end of an axon into the synaptic cleft. The neurotransmitters bind to receptors on the postsynaptic terminal, which may be a neuron or a muscle cell in the case of a neuromuscular junction. These are collectively referred to as postsynaptic receptors, since they are on the membrane of the postsynaptic cell. Neurotransmitters bind to their receptors by having a particular shape or structure, somewhat like the way a key fits into certain locks. Postsynaptic potentials are subject to summation, spatially and/or temporally. Excitatory postsynaptic potential (EPSP) is a temporary depolarization of postsynaptic membrane potential caused by the flow of positively charged ions into the postsynaptic cell as a result of opening of ligand-sensitive channels. A postsynaptic potential is defined as excitatory if it makes it easier for the neuron to fire an action potential. IPSPs are sometimes caused by an increase in positive charge outflow. The flow of ions that causes an EPSP is an excitatory postsynaptic current (EPSC). Inhibitory postsynaptic postsynaptic potentials. An inhibitory postsynaptic potential (IPSP) is a synaptic potential that decreases the chance that a future action potential will occur in a postsynaptic neuron or α-motoneuron.. They can take place at all chemical synapses which utilize the secretion
  • 39. of neurotransmitters to create cell to cell signaling. Inhibitory presynaptic neurons release neurotransmitters which then bind to the postsynaptic receptors; this induces a postsynaptic conductance change as ion channels open or close. An electrical current is generated which changes the postsynaptic membrane potential to create a more negative postsynaptic potential. Depolarization can also occur due to an IPSP if the reverse potential is between the resting threshold and the action potential threshold. Another way to look at inhibitory postsynaptic potentials is that they are also a chlorine conductance change in the neuronal cell because it decreases the driving force. In general, a postsynaptic potential is dependent on the type and combination of receptor channel, reverse potential of the postsynaptic potential, action potential threshold voltage, ionic permeability of the ion channel, as well as the concentrations of the ions in and out of the cell; this determines if it is excitatory or inhibitory. IPSPs always want to keep the membrane potential more negative than the action potential threshold and can be seen as a “transient hyperpolarization” EPSPs and IPSPs compete with each other at numerous synapses of a neuron; this determines whether or not the action potential at the presynaptic terminal will regenerate at the postsynaptic membrane. Some common neurotransmitters involved in IPSPs are GABA and glycine. 29 – Describe structural arrangement of the neuromuscular junction and its function. The neuromuscular junction: The skeletal muscle fibers are innervated by large, myelinated nerve fibers that originate from large motorneurons in the anterior horns of the spinal cord. Each nerve fiber, after entering the muscle belly, normally branches and stimulates from three to several hundred several skeletal muscle fibers. Each nereve endings makes a junction, neuromuscular junction, with the muscle fiber near its midpoint. The action potential initiated in the muscle fiber by the nerve signal travels in both directions towards the muscle fiber ends. The nerve fibres forms a complex of branching nerve terminals that invaginate into the surface os the muscle fiber but lie outside the muscle fiber plasma membrane. The entire structure is called the motor end plate. It is vovered by one or more Scwmann cells that insulate it from the surronfing fluids. In the junction between a single axon terminal and the muscle fiber membrane, the invaginated membrane is called the synaptic gutter or synaptic trough, and the space between the terminal and the fiber membrane is called the synaptic space or synaptic cleft. This space is 20 to 30 nanometers wide. At the bottom of the gutter are numerous smaller folds of the muscle membrane called subneural clefts, which gratly increase the surface area at which the synaptic transmitter can act. In the axon terminal are many mitochondria that supply ATP, the energy source that is used for the synthesis of an excitatory transmitter acetylcholine. The acetylcholine in turn excites the muscle fiber membrane. Acetylcholine is synthetized in the cytoplasm of the terminal, about 300.000 of which are normally in the terminals of a single end plate. In the synaptic space are large quantities of the enzyme acetylcholinesterase, which destroys acetylcholine a few milliseconds after it has been released from the synaptic vesicles. When a nerve impulse reaches the neuromuscular junction, about 125 vesicles of acetylcholine are released from the terminals into synaptic space.
  • 40. On the inside surface of the neural membrane are linear dense bars. To each side of each dense bar are protein particles that penetrate the neural membrane; these are voltage-gated calcium channels. When an action potential spreads over the terminal, these channels open and allow calcium ions to diffuse from the synaptic space to the interior of the nerve terminal. The calcium ions, in turn, are believed to exert an attractive influence on the acetylcholine vesicles, drawing them to the neural membrane adjacent to dense bars. The vesicles then fuse with the neural membrane and empty their acetylcholine into the synaptic space by the process of exocytosis. Myastenia gravis and Lambert-Eaton syndrome. Myastenis gravis is a serious and sometimes fatal disease in which skeletal muscles are weak. It is caused by the formation of circulatory antibodies to the muscle type of nicotinic acetylcholine receptors. These antibodies destroy some of the receptors and bind others to neighbouring receptors triggering their removal by endocytosis. Drugs affecting transmission at neuromuscular junction: a) some drugs act on acetylcholine thus stimulating the muscle fiber, e.g. nicotine b) some drugs may block transmission e.g. muscle relaxant. c) some drugs stimulate neuromuscular junction by inactivation ach- acetylcholinesterase e.g. physostigmine 30 – Presynaptic inhibition and posttetanic potentiation In addition to the inhibition caused by inhibitory synapses operating at the neuronal membranes which is called post-synaptic inhibition, another type of inhibition often ocuurs in the presynaptic terminals before the signals even reaches the synapse – pre-synaptic inhibition. Pre-synaptic inhibition is the phenomenon that occurs when a presynaptic neuron exerts inhibitory influences through transmitters at an axo-axonic synapse with the terminal in a postsynaptic neuron. In presynaptic inhibition, the inhibition is caused by discharge of inhibitory synapses that lie on the presynaptic terminals nerve fibrils before their endings terminate on the post-synaptic neuron. In most instances, the inhibitory transmitter released is GABA, this has the specific effect of opening ion channels, allowing CL- to diffuse into the terminal fibril. Therefore the action potential itself becomes greatly reduced, thus also reducing the degree of excitation of the postsynaptic neuron. Presynaptic inhibition occurs in many of the sensory pathways in the nervous system. 3 mechanisms of presynaptic inhibition are seen: ⇒ Activation of the presynaptic receptors increases Cl- conductance, and this has been shown to decrease the size of the action potentials rwaching the excitatory ending. ⇒ This in turn reduces Ca++ entry and consequently the amount of excitatory transmitter released. Voltage-gated K+ channels are also opened and the resulting K+ also decreases Ca++ influx. ⇒ Evidence for direct inhibition of transmitter release independent of Ca++ influx into the excitatory ending is evident.
  • 41. Post-tetanic potentiation If we stimulate motor unit and correlate it with the srenght of muscle contraction we can observe the principles of frequency summation and tetanization. First, with a lower rate of stimulation (times per second) and then increasing the frequency there comes a point where each new contraction occurs before the preeciding one is over. As a result, the second contraction is added partially to the first, so that the total strength of contraction rises progressively with increasing frequency. When the frequency reaches a critical level, the successive contractions eventually become so rapid that they fuse together, and the whole muscle contraction appears to be completely smooth and continuos. This is called tetanization. At a slightly higher frequency, the strength of contraction reaches its maximum, so that any additional increase in frequency beyond that point has no further effect in increasing contractile force. This occurs because enough calcium ions are maintained in the muscle sarcoplasm, even between action potentials, so that full contractile state is sustained without allowing any relaxation between the action potentials. The tetanizing stimulation causes Ca++ to accumulate in the presynaptic neuron to such a degree that the intracellular binding sites that keep cytoplasm Ca++ low are overwhelmed. Postetanic potentiation is opposite to habituation 31 – Spontaneously active neurons: Many neurons do not maintain a steady resting potential but fire impulses spontaneously. Two patterns often seen are: a) regular firing or “beating” b) grouped firing or “bursting” In spontaneously active neurons, resting Na+ conductance is high, the leakage current depolarizes and the threshold is crossed, and an action potential is fired. a) In regular firing neurons: in these neurons the membrane potential is less negative due to their possession of high Na+ leak conductance and low K+ leak conductancy. As the membrane potential moves to a more positive value, the thereshold is crossed quickly. After an impulse, voltage K+ channels open and the membrane is hyperepolarized. There is then a return to the resting potential due to to Na+ leakage. b)In bursting neurons: these neurons have the same membrane channels as firing neurons, they regulate action potentials during a burst. In depolarization of an action potential, voltage-gated Ca++ channels are opened to allow Ca++. They are slow, voltage-gated channels allow Na+, Ca++ exchange. This slow depolarization predominate during the impulse generating phase of the burst cycle. K+ channels which are activated by Ca++ are responsible for hyperrepolarisation (calcium-gated K+ channel). In bursting neurons, the leakage is slow, voltage dependent depolarizing channels (Na+, Ca++) generate the burst in a action potential. During this phase, Ca++ enters the cell by the voltage dependent Ca++ channels and activates Ca++
  • 42. gated K+ channels. K + hyperrepolarizes the membrane until the moment when Ca++ is pumped out of the cell or sequestrated. Spontaneous active neurons are important for circuits controlling respiration, locomoytion as well as other activities 32 – Coding of sensory information Cells specialized to respond to a specific environmental stimuli are termed sensory receptor cells. a) a neuron specialized for sensory reception b) a receptor cell connected to an afferent neuron c) sense organ composed of receptor cells and additional accessory to transform the stimulus. Receptor cells respond specifically to certain stimuli (receptor specificity) , there are 4 major groups of receptors in mammals: a) thermoreceptors b) chemoreceptors c) photoreceptors d) mechanoreceptors A stimulus is characterized by its modality, intensity, duration and location. Sensation evoked by a stimulus depends on the part of the brain that has been stimulated – Pacinian corpuscle stimulation either by touch or imitation from a tumour produces a touch sensation. Sensory information are processed by the thalamus and is transmitted to the cerebral cortex where the nerve pathways from a particular sense organ are stimulated, the sensation evoked is that for which the receptor is specialized, no matter how long the pathway or where along the pathway the activity has been initiated. Law of projection: no matter where a particular sensory pathway is stimulated along its course to the cortex, the conscious adaptation produced is referred to the location of the receptor, (e.g. phantom limb sensation seen in amputated people or in neurosurgical experiments with conscious patients, stimulation of specific part of cortex may produce tickling sensations)