Ynaptic Ransmission: The Human Brain Contains at Least

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 32

5

Synaptic Transmission

Overview
THE HUMAN BRAIN CONTAINS AT LEAST 100 billion neurons, each with the
ability to influence many other cells. Clearly, sophisticated and highly efficient mech-
anisms are needed to enable communication among this astronomical number of
elements. Such communication is made possible by synapses, the functional contacts
between neurons. Two different types of synapses—electrical and chemical—can be
distinguished on the basis of their mechanism of transmission. At electrical synapses,
current flows through gap junctions, which are specialized membrane channels that
connect two cells. In contrast, chemical synapses enable cell-to-cell communication
via the secretion of neurotransmitters; these chemical agents released by the presyn-
aptic neurons produce secondary current flow in postsynaptic neurons by activating
specific receptor molecules. The total number of neurotransmitters is well over 100.
Virtually all neurotransmitters undergo a similar cycle of use: synthesis and packag-
ing into synaptic vesicles; release from the presynaptic cell; binding to postsynaptic
receptors; and, finally, rapid removal and/or degradation. The influx of Ca2+ through
voltage-gated channels triggers the secretion of neurotransmitters; this, in turn, gives
rise to a transient increase in Ca2+ concentration within the presynaptic terminal. The
rise in Ca2+ concentration causes synaptic vesicles to fuse with the presynaptic plasma
membrane and release their contents into the space between the pre- and postsyn-
aptic cells. Although it is not yet understood exactly how Ca2+ triggers exocytosis,
specific proteins on the surface of the synaptic vesicle and the presynaptic plasma
membrane mediate this process. Neurotransmitters evoke postsynaptic electrical
responses by binding to members of a diverse group of neurotransmitter receptors.
There are two major classes of receptors: those in which the receptor molecule is
also an ion channel, and those in which the receptor and ion channel are separate
entities. These receptors give rise to electrical signals by transmitter-induced open-
ing or closing of the ion channels. Whether the postsynaptic actions of a particular
neurotransmitter are excitatory or inhibitory is determined by the ionic permeability
of the ion channel affected by the transmitter, and by the electrochemical gradient
for the peremeant ions.

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
78  C h a p t e r 5

Connexons are present in the membranes of both the pre-


Electrical Synapses and postsynaptic neurons; six presynaptic connexins align
The many kinds of synapses within the human brain fall with six postsynaptic connexins to form a pore that con-
into two general classes: electrical synapses and chemical nects the two cells (Figure 5.1C). The pore of a connexon
synapses. Although they are a distinct minority, electri- channel is much larger than the pores of the voltage-gated
cal synapses are found in all nervous systems and permit ion channels described in the previous chapter. As a result,
direct, passive flow of electrical current from one neuron
to another. The structure of an electrical synapse is shown FIGURE 5.1  Structure of electrical synapses. (A) At electri-
schematically in Figure 5.1. The “upstream” neuron, which cal synapses, gap junctions occur between pre- and postsynaptic
is the source of current, is called the presynaptic element, membranes. (B) Gap junctions consist of intercellular channels
and the “downstream” neuron into which this current that permit current to flow passively from the presynaptic cell
flows is termed postsynaptic. The membranes of the two to the postsynaptic cell (C) Gap junctions consist of hexameric
communicating neurons come extremely close at the syn- complexes formed by the coming together of subunits called
apse and are actually linked together by an intercellular connexons, which are present in both the pre- and postsynaptic
specialization called a gap junction. Gap junctions con- membranes. The pores of the channels connect, creating electri-
tain precisely aligned, paired channels called connexons. cal continuity between the two cells. (D) Connexons consist of
the integral membrane protein connexin.

(A) (B)
Presynaptic Ionic current flows through
Microtubules connexon channels
membrane
Cytoplasm
Presynaptic
neuron Mitochondrion

Extracellular

Postsynaptic
Postsynaptic membrane Connexons
Gap junction neuron

(C) Presynaptic (D)


membrane Extracellular

Connexons Connexin

Postsynaptic
3.5 nm
membrane 20 nm

Pores connecting
cytoplasm of two
neurons

Cytoplasm C

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   79

a variety of substances can simply diffuse between the cyto- types of connexins, found in different cell types and yield-
plasm of the pre- and postsynaptic neurons. In addition to ing gap junctions with diverse physiological properties.
ions, substances that diffuse through gap junction pores in- Electrical synapses work by allowing ionic current to flow
clude molecules with molecular weights as great as several passively through the gap junction pores from one neuron
hundred daltons. This permits ATP and other important to another. The usual source of this current is the poten-
intracellular metabolites, such as second messengers (see tial difference generated locally by the presynaptic action
Chapter 7), to be transferred between neurons. Connexons potential (see Chapter 3). This arrangement has a number
are composed of a special family of ion channel proteins, of interesting consequences. One is that transmission can
the connexins (Figure 5.1D). There are several different be bidirectional; that is, current can flow in either direction
across the gap junction, depending on which member of
(A)
the coupled pair is invaded by an action potential (although
some types of gap junctions have special features that render
their transmission unidirectional). Another important feature
25 Presynaptic
neuron of the electrical synapse is that transmission is extraordinarily
fast: because passive current flow across the gap junction is
0
virtually instantaneous, communication can occur without
the delay that is characteristic of chemical synapses.
−25
Membrane potential (mV)

These features are apparent in the operation of the first


−50 electrical synapse to be discovered, which resides in the
crayfish nervous system. A postsynaptic electrical signal is
observed at this synapse within a fraction of a millisecond
after the generation of a presynaptic action potential (Fig-
ure 5.2A). In fact, at least part of this brief synaptic delay
25 is caused by propagation of the action potential into the
Postsynaptic presynaptic terminal, so that there may be essentially no
0 neuron
delay at all in the transmission of electrical signals across
−25
the synapse. Such synapses interconnect many of the neu-
rons within the circuit that allows the crayfish to escape
−50 from its predators, thus minimizing the time between the
Minimal (<0.1 ms)
synaptic delay
presence of a threatening stimulus and a potentially lifesav-
ing motor response.
0 1 2 3 4
A more general purpose of electrical synapses is
Time (ms)
(B)
to synchronize electrical activity among populations
of neurons. For example, the brainstem neurons
0 that generate rhythmic electrical activity underlying
breathing are synchronized by electrical synapses, as
are populations of interneurons in the cerebral cor-
−25
tex, thalamus, cerebellum, and other brain regions
(Figure 5.2B). Electrical transmission between certain
Membrane potential (mV)

−50 hormone-secreting neurons within the hypothalamus


ensures that all cells fire action potentials at about
Neuron 1
the same time, thus facilitating a burst of hormone

* * * * * * *
0 FIGURE 5.2  Function of gap junctions at electrical
synapses. (A) Rapid transmission of signals at an electrical
synapse in the crayfish. An action potential in the presynap-
−25 tic neuron causes the postsynaptic neuron to be depolar-
ized within a fraction of a millisecond. (B) Electrical synapses
−50 allow synchronization of electrical activity in hippocampal
interneurons. In a pair of interneurons connected by elec-
Neuron 2 trical synapses, generation of an action potential in one
neuron often results in the synchronized firing of an action
0 100 200 300 400 potential in another neuron. (A after Furshpan and Potter,
Time (ms) 1959; B after Beierlein et al., 2000.)

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
80  C h a p t e r 5

secretion into the circulation. The fact that gap junction synapses than at electrical synapses and is called the synap-
pores are large enough to allow molecules such as ATP and tic cleft. However, the key feature of all chemical synapses
second messengers to diffuse between cells also permits is the presence of small, membrane-bounded organelles
electrical synapses to coordinate the intracellular signaling called synaptic vesicles within the presynaptic terminal.
and metabolism of coupled cells. This property may be par- These spherical organelles are filled with one or more neu-
ticularly important for glial cells, which form large intracel- rotransmitters, the chemical signals secreted from the pre-
lular signaling networks via their gap junctions. synaptic neuron, and it is these chemical agents acting as
messengers between the communicating neurons that gives
Signal Transmission at this type of synapse its name.
Chemical Synapses Transmission at chemical synapses is based on the elab-
orate sequence of events depicted in Figure 5.3. The process
The general structure of a chemical synapse is shown sche-
matically in Figure 5.3. The space between the pre- and
postsynaptic neurons is substantially greater at chemical
FIGURE 5.3  Sequence of events involved in transmission
at a typical chemical synapse.

Myelin

An action potential invades


1 Transmitter is the presynaptic terminal
synthesized and then
stored in vesicles
3 Depolarization of presynaptic
terminal causes opening of
voltage-gated Ca2+ channels

2+
4 Influx of Ca
through channels
Synaptic
vesicle 2+
5 Ca causes vesicles to
11 Retrieval of vesicular fuse with presynaptic
membrane from membrane
plasma membrane Transmitter
molecules Ca2+
6 Transmitter is released
into synaptic cleft
Glial cell via exocytosis

Across
dendrite
Trans-
mitter
molecules

Postsynaptic
current flow
Ions
Transmitter
receptor
7 Transmitter binds to
10 Removal of neuro- receptor molecules in
transmitter by glial postsynaptic membrane
uptake or enzymatic 9 Postsynaptic current causes 8 Opening or closing
degradation excitatory or inhibitory of postsynaptic
postsynaptic potential that channels
changes the excitability of
the postsynaptic cell

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   81

is initiated when an action potential invades the terminal


of the presynaptic neuron. The change in membrane po-
Properties of Neurotransmitters
tential caused by the arrival of the action potential leads The notion that electrical information can be transferred
to the opening of voltage-gated calcium channels in the from one neuron to the next by means of chemical signal-
presynaptic membrane. Because of the steep concentration ing was the subject of intense debate through the first half
gradient of Ca2+ across the presynaptic membrane (the ex- of the twentieth century. In 1926, the German physiologist
ternal Ca2+ concentration is approximately 10–3 M, whereas Otto Loewi performed a key experiment that supported this
the internal Ca2+ concentration is approximately 10–7 M), idea. Acting on an idea that allegedly came to him in the
the opening of these channels causes a rapid influx of Ca2+ middle of the night, Loewi proved that electrical stimula-
into the presynaptic terminal, with the result that the Ca2+ tion of the vagus nerve slows the heartbeat by releasing a
concentration of the cytoplasm in the terminal transiently chemical signal. He isolated and perfused the hearts of two
rises to a much higher value. Elevation of the presynaptic frogs, monitoring the rates at which they were beating (Fig-
Ca2+ concentration, in turn, allows synaptic vesicles to fuse ure 5.4). When the vagus nerve innervating the first heart
with the plasma membrane of the presynaptic neuron. The was stimulated, the beat of this heart slowed. Remarkably,
Ca2+-dependent fusion of synaptic vesicles with the termi- even though the vagus nerve of the second heart had not
nal membrane causes their contents, most importantly neu- been stimulated, its beat also slowed when exposed to the
rotransmitters, to be released into the synaptic cleft. perfusate from the first heart. This result showed that the
Following exocytosis, transmitters diffuse across the syn- vagus nerve regulates the heart rate by releasing a chemi-
aptic cleft and bind to specific receptors on the membrane cal that accumulates in the perfusate. Originally referred
of the postsynaptic neuron. The binding of neurotransmit- to as “vagus substance,” the agent was later shown to be
ter to the receptors causes channels in the postsynaptic acetylcholine (ACh). ACh is now known to be a neu-
membrane to open (or sometimes to close), thus changing rotransmitter that acts not only in the heart but also at a
the ability of ions to flow into (or out of) the postsynaptic variety of postsynaptic targets in the central and peripheral
cells. The resulting neurotransmitter-induced current flow nervous systems, preeminently at the neuromuscular junc-
alters the conductance and (usually) the membrane poten- tion of striated muscles and in the visceral motor system
tial of the postsynaptic neuron, increasing or decreasing (see Chapters 6 and 21).
the probability that the neuron will fire an action potential. Over the years, a number of formal criteria have emerged
Subsequent removal of the neurotransmitter from the syn- that definitively identify a substance as a neurotransmitter
aptic cleft, by uptake into glial cells or by enzymatic deg-
radation, terminates the action of the neurotransmitter. In
this way, information is transmitted transiently from one
FIGURE 5.4  Loewi’s experiment demonstrated chemical
neuron to another.
neurotransmission. (A) Loewi’s experimental setup. (B) Where
the vagus nerve of an isolated frog’s heart was stimulated, the
heart rate decreased (upper panel). If the perfusion fluid from this
(A) (B) Stimulate vagus heart was transferred to a second heart, the second heart’s rate
Stimulate decreased as well (lower panel).
nerve of heart 1

Vagus Heart 1
Heartbeat
nerve slowed
Contraction force

Solution
transferred
to heart 2

Time (s)
Heart 1
Heart 2
Inhibitory effect
of vagus transferred
Contraction force

Heart 2
Time (s)

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
82  C h a p t e r 5

(Box 5A). These have led to the identification of more than FIGURE 5.5  Metabolism of small-molecule and peptide

t
100 different neurotransmitters, which can be classified into transmitters. (A) Small-molecule neurotransmitters are synthe-
two broad categories: small-molecule neurotransmitters and sized at nerve terminals. The enzymes necessary for neurotrans-
neuropeptides (see Chapter 6). Having more than one trans- mitter synthesis are made in the cell body of the presynaptic
mitter diversifies the physiological repertoire of synapses. cell (1) and are transported down the axon by slow axonal trans-
port (2). Precursors are taken up into the terminals by specific
Multiple neurotransmitters can produce different types of
transporters, and neurotransmitter synthesis and packaging
responses on individual postsynaptic cells. For example, a
take place within the nerve endings (3). After vesicle fusion and
neuron can be excited by one type of neurotransmitter and release (4), the neurotransmitter may be enzymatically degraded.
inhibited by another type of neurotransmitter. The speed of The reuptake of the neurotransmitter (or its metabolites) begins
postsynaptic responses produced by different transmitters another cycle of synthesis, packaging, release, and removal (5).
also differs, allowing control of electrical signaling over dif- (B) Small clear-core vesicles at a synapse between an axon ter-
ferent time scales. In general, small-molecule neurotransmit- minal (AT) and a dendritic spine (Den) in the central nervous sys-
ters mediate rapid synaptic actions, whereas neuropeptides tem. Such vesicles typically contain small-molecule neurotrans-
tend to modulate slower, ongoing synaptic functions. mitters. (C) Peptide neurotransmitters, as well as the enzymes
Until relatively recently, it was believed that a given that modify their precursors, are synthesized in the cell body
neuron produced only a single type of neurotransmitter. (1). Enzymes and propeptides are packaged into vesicles in the
Golgi apparatus. During fast axonal transport of these vesicles to
It is now clear, however, that many types of neurons syn-
the nerve terminals (2), the enzymes modify the propeptides to
thesize and release two or more different neurotransmit-
produce one or more neurotransmitter peptides (3). After vesicle
ters. When more than one transmitter is present within a fusion and exocytosis, the peptides diffuse away and are degrad-
nerve terminal, the molecules are called co-transmitters. ed by proteolytic enzymes (4). (D) Large dense-core vesicles in a
Because different types of transmitters can be packaged in central axon terminal (AT) synapsing onto a dendrite (Den). Such
different populations of synaptic vesicles, co-transmitters vesicles typically contain neuropeptides or, in some cases, bio-
need not be released simultaneously. When peptide and genic amines. (B and D from Peters, Palay, and Webster, 1991.)
small-molecule neurotransmitters act as co-transmitters at
the same synapse, they are differentially released according
to the pattern of synaptic activity: low-frequency activity these vesicles are referred to as small clear-core vesicles
often results in the release of only small neurotransmitters, (Figure 5.5B). Neuropeptides are synthesized in the cell
whereas high-frequency activity is required to release neu- body of a neuron, meaning that the peptide is produced a
ropeptides from the same presynaptic terminals. As a result, long distance away from its site of secretion (Figure 5.5C).
the chemical signaling properties of such synapses change To solve this problem, peptide-filled vesicles are transported
according to the rate of activity. along an axon and down to the synaptic terminal via fast
Effective synaptic transmission requires close control of axonal transport. This process carries vesicles at rates up to
the concentration of neurotransmitters within the synap- 400 mm/day along cytoskeletal elements called microtubules
tic cleft. Neurons have therefore developed a sophisticated (in contrast with the slow axonal transport of the enzymes
ability to regulate the synthesis, packaging, release, and that synthesize small-molecule transmitters). Microtubules
degradation (or removal) of neurotransmitters to achieve are long, cylindrical filaments, 25 nm in diameter, present
the desired levels of transmitter molecules. The synthesis throughout neurons and other cells. Peptide-containing
of small-molecule neurotransmitters occurs locally within vesicles move along these microtubule “tracks” by ATP-
presynaptic terminals (Figure 5.5A). The enzymes needed requiring “motor” proteins such as kinesin. Neuropeptides
to synthesize these transmitters are produced in the neu- are packaged into synaptic vesicles that range from 90–250
ronal cell body and are then transported to the nerve ter- nm in diameter. Because the center of these vesicles appear
minal cytoplasm at a rate of 0.5–5.0 millimeters a day by electron-dense in electron micrographs, they are referred to
a mechanism called slow axonal transport. The precursor as large dense-core vesicles (Figure 5.5D).
molecules required to make new molecules of neurotrans- After a neurotransmitter has been secreted into the syn-
mitter are usually taken into the nerve terminal by trans- aptic cleft, it must be removed to enable the postsynaptic
porters found in the plasma membrane of the terminal. The cell to engage in another cycle of synaptic transmission. The
enzymes synthesize neurotransmitters in the cytoplasm removal of neurotransmitters involves diffusion away from
of the presynaptic terminal, and the transmitters are then the postsynaptic receptors, in combination with reuptake
loaded into synaptic vesicles via transporters in the vesicular into nerve terminals or surrounding glial cells, degradation
membrane (see Chapter 4). For some small-molecule neu- by specific enzymes, or a combination of these mechanisms.
rotransmitters, the final steps of synthesis occur inside the Specific transporter proteins remove most small-molecule
synaptic vesicles. Most small-molecule neurotransmitters neurotransmitters (or their metabolites) from the synaptic
are packaged in vesicles 40–60 nm in diameter, the centers cleft, ultimately delivering them back to the presynaptic ter-
of which appear clear in electron micrographs; accordingly, minal for reuse (see Chapter 6).

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
(A) Small-molecule transmitter (C) Peptide transmitter

Nucleus 1 Synthesis 1 Synthesis of


of enzymes neurotransmitter
in cell body precursors and
enzymes

RER

Golgi
apparatus

Microtubules

2 Transport of enzymes
and peptide
precursors down
2 Slow axonal microtubule tracks
transport
of enzymes

Axon

5 Transport
of precursors
into terminal
Terminal

4 Neurotransmitter 3 Enzymes modify


Precursor Enzymes 3 Synthesis and diffuses away and precursors to
packaging of is degraded by produce peptide
neurotransmitter proteolytic enzymes neurotransmitter
4 Release and
diffusion
of neuro–
Neuro-
transmitter
transmitter

(B) (D)

Presynaptic
terminals

Vesicles

Dendrites

0.5 µm

PURVES: Neuroscience 5e
Copyright
Figure: 05.05 © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
07/20/11 in any form without express written permission from the publisher.
07/26/11
84  C h a p t e r 5

Box 5A  Criteria That Define a Neurotransmitter


Three primary criteria have been used in all neurons, their presence is not 3. Specific receptors for the substance must
to confirm that a molecule acts as a sufficient evidence to establish them be present on the postsynaptic cell. A
neurotransmitter at a given chemical as neurotransmitters. neurotransmitter cannot act on its
synapse. 2. The substance must be released in target unless specific receptors for
1. The substance must be present within response to presynaptic depolarization, the transmitter are present in the
the presynaptic neuron. Clearly, a and the release must be Ca2+-depen- postsynaptic membrane. One way to
chemical cannot be secreted from dent. Another essential criterion for demonstrate the presence of recep-
a presynaptic neuron unless it is identifying a neurotransmitter is to tors is to show that application of
present there. Because elaborate demonstrate that it is released from exogenous transmitter mimics the
biochemical pathways are required the presynaptic neuron in response postsynaptic effect of presynaptic
to produce neurotransmitters, show- to presynaptic electrical activity, stimulation. A more rigorous dem-
ing that the enzymes and precursors and that this release requires Ca2+ onstration is to show that agonists
required to synthesize the substance influx into the presynaptic terminal. and antagonists that alter the normal
are present in presynaptic neurons Meeting this criterion is technically postsynaptic response have the same
provides additional evidence that the challenging, not only because it may effect when the substance in question
substance is used as a transmitter. be difficult to selectively stimulate is applied exogenously. High-reso-
Note, however, that since the trans- the presynaptic neurons, but also lution histological methods can also
mitters glutamate, glycine, and aspar- because enzymes and transport- be used to show that specific recep-
tate are also needed for protein syn- ers efficiently remove the secreted tors are present in the postsynaptic
thesis and other metabolic reactions neurotransmitters. membrane (by detection of radioac-
tively labeled receptor antibodies, for
example).
(1) (2) (3)
Fulfilling these criteria establishes
unambiguously that a substance is used
as a transmitter at a given synapse.
1 Neurotransmitter Practical difficulties, however, have
present Action
potential prevented these standards from being
Presynaptic
terminal
applied at many types of synapses. It is
Application for this reason that so many substances
of transmitter,
+ agonists, or
must be referred to as “putative”
Ca2 neurotransmitters.
+ antagonists
Ca2

Demonstrating the identity of a neuro-


transmitter at a synapse requires show-
Postsynaptic cell
ing (1) its presence, (2) its release, and
2 Neurotransmitter 3 Neurotransmitter (3) the postsynaptic presence of specific
released receptors activated receptors.

University College London, and Katz has been widely rec-


Quantal Release of Neurotransmitters ognized for his remarkable contributions to understanding
Much of the evidence leading to the present understanding synaptic transmission. Though he worked primarily on the
PURVES: Neuroscience 5e
of chemical synaptic transmission was obtained from ex-
Figure: BX05A.A frog neuromuscular junction, numerous subsequent experi-
periments
07/20/11examining the release of ACh at neuromuscular ments have confirmed the applicability of his observations
07/22/11These synapses between spinal motor neurons
junctions. to transmission at all chemical synapses.
and skeletal muscle cells are simple, large, and peripherally When an intracellular microelectrode is used to record
located, making them particularly amenable to experimen- the membrane potential of a muscle cell, an action potential
tal analysis. Such synapses occur at specializations called in the presynaptic motor neuron can be seen to elicit a tran-
end plates because of the saucer-like appearance of the site sient depolarization of the postsynaptic muscle fiber. This
on the muscle fiber where the presynaptic axon elaborates change in membrane potential, called an end plate poten-
its terminals (Figure 5.6A). Most of the pioneering work tial (EPP), is normally large enough to bring the membrane
on neuromuscular transmission was performed during the potential of the muscle cell well above the threshold for
1950s and 1960s by Bernard Katz and his collaborators at producing a postsynaptic action potential (Figure 5.6B). The

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   85
(A) Stimulate Stimulate
axon

Record FIGURE 5.6  Synaptic transmission at the neuromuscular junction. (A) Experimental
arrangement: the axon of the motor neuron innervating the muscle fiber is stimulated with an
Record
postsynaptic extracellular electrode, while an intracellular microelectrode is inserted into the postsynaptic
Axon muscle cell to record its electrical responses. (B) End plate potentials (shaded area) evoked by
membrane
potential stimulation of a motor neuron are normally above threshold and therefore produce an action
potential in the postsynaptic muscle cell. (C) Spontaneous miniature EPPs (MEPPs) occur in
the absence of presynaptic stimulation. (D) When the neuromuscular junction is bathed in
a solution that has a low concentration of Ca2+, stimulating the motor neuron evokes EPPs
Muscle cell whose amplitudes are reduced to about the size of MEPPs. (After Fatt and Katz, 1952.)

(B) Stimulate (C) (D) Stimulate


motor axon motor axon
+50
Postsynaptic membrane

Postsynaptic membrane

Postsynaptic membrane
Action Subthreshold EPP
1 mV 1 mV
potential MEPP
potential (mV)

potential (mV)

potential (mV)
0
Threshold
−50

−100
End plate Spontaneous
potential (EPP) MEPP
0 2 4 6 0 200 400 0 20 40 60 80 100
Time (ms) Time (ms) Time (ms)

postsynaptic action potential triggered by the EPP causes potential production and allowing it to be measured more
the muscle fiber to contract. Unlike the case for electrical precisely. Under such conditions, stimulation of the motor
synapses, there is a pronounced delay between the time neuron produces very small EPPs that fluctuate in ampli-
that the presynaptic motor neuron is stimulated and when tude from trial to trial (Figure 5.6D). These fluctuations give
the EPP occurs in the postsynaptic muscle cell. Such a delay considerable insight into the mechanisms responsible for
is characteristic of all chemical synapses. neurotransmitter release. In particular, the variable evoked
One of Katz’s seminal findings, in studies carried out response in low Ca2+ is now known to result from the release
with Paul Fatt in 1951, was that spontaneous changes in of unit amounts of ACh by the presynaptic nerve terminal.
muscle cell membrane potential occur even in the absence Indeed, the amplitude of the smallest evoked response
of stimulation of the presynaptic motor neuron (Figure is strikingly similar to the size of single MEPPs (compare
5.6C). These changes have the same shape as EPPs but are Figures 5.6C and D). Further supporting this similarity, in-
much smaller (typically less than 1 mV in amplitude, com- crements in the EPP response (Figure 5.7A) occur in units
pared
PURVES:with an EPP of
Neuroscience 5e more than 50 mV). Both EPPs and about the size of single MEPPs (Figure 5.7B). These “quan-
Figure: 05.06
these small, spontaneous events are sensitive to pharmaco-
07/18/11
tal” fluctuations in the amplitude of EPPs indicated to Katz
logical
09/23/11agents that block postsynaptic acetylcholine recep- and his colleague, Jose del Castillo, that EPPs are made up
tors, such as curare (see Box 6B). These and other parallels of individual units, each equivalent to a MEPP.
between EPPs and the spontaneously occurring depolariza- The idea that EPPs represent the simultaneous release of
tions led Katz and his colleagues to call these spontaneous many MEPP-like units can be tested statistically. A method
events miniature end plate potentials, or MEPPs. of statistical analysis based on the independent occurrence
The relationship between the full-blown end plate po- of unitary events (called Poisson statistics) predicts what
tential and MEPPs was clarified by careful analysis of the the distribution of EPP amplitudes would look like during
EPPs. The magnitude of the EPP provides a convenient a large number of trials of motor neuron stimulation, under
electrical assay of neurotransmitter secretion from a motor the assumption that EPPs are built up from unitary events
neuron terminal; however, measuring it is complicated by represented by MEPPs (see Figure 5.7B). The distribution
the need to prevent muscle contraction from dislodging the of EPP amplitudes determined experimentally was found
microelectrode. The usual means of eliminating muscle con- to be just that expected if transmitter release from the mo-
tractions is either to lower Ca2+ concentration in the extra- tor neuron is indeed quantal (the red curve in Figure 5.7A).
cellular medium or to partially block the postsynaptic ACh Such analyses confirmed the idea that release of acetylcho-
receptors with the drug curare. As expected from the scheme line does indeed occur in discrete packets, each equivalent
illustrated in Figure 5.3, lowering the Ca2+ concentration re- to a MEPP. In short, a presynaptic action potential causes
duces neurotransmitter secretion, thus reducing the magni- a postsynaptic EPP because it synchronizes the release of
tude of the EPP below the threshold for postsynaptic action many transmitter quanta.

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
86  C h a p t e r 5

(A) No EPP in vesicle (∼50 nm), approximately 10,000 molecules of


response to neurotransmitter are contained in a single vesicle. This
stimulation
Prediction of
number corresponds quite nicely to the amount of ACh
20 that must be applied to a neuromuscular junction to
statistical model
mimic a MEPP, providing further support for the idea
that quanta arise from discharge of the contents of sin-
Number of EPPs

15 gle synaptic vesicles.


To prove that quanta are caused by the fusion of
individual synaptic vesicles with the plasma membrane,
10 it is necessary to show that each fused vesicle causes a
single quantal event to be recorded postsynaptically.
This challenge was met in the late 1970s, when John
5
Heuser, Tom Reese, and colleagues correlated mea-
surements of vesicle fusion with the quantal content of
EPPs at the neuromuscular junction. They used elec-
tron microscopy to determine the number of vesicles
0
0 0.4 0.8 1.2 1.6 2.0 2.4 2.8 that fused with the presynaptic plasma membranes of
EPP amplitude (mV) terminals that had been treated with a drug (4-amino-
pyridine, or 4-AP) that enhances the number of quanta
(B) FIGURE 5.7  Quantized distribution of released by single action potentials (Figure 5.8A). Par-
EPP amplitudes evoked in a low-Ca2+ allel electrical measurements were made of the quantal
30 solution. Peaks of EPP amplitudes (A) tend content of the EPPs elicited in this way. A comparison
to occur in integer multiples of the mean of the number of synaptic vesicle fusions observed with
the electron microscope and the number of quanta re-
Number of MEPPs

amplitude of MEPPs, whose amplitude dis-


tribution is shown in (B). The leftmost bar leased at the synapse showed a good correlation be-
20 in the EPP amplitude distribution shows tween these two measures (Figure 5.8B). These results
trials in which presynaptic stimulation remain one of the strongest lines of support for the idea
failed to elicit an EPP in the muscle cell. that a quantum of transmitter release is due to fusion
The red curve indicates the prediction of a
10 of a single synaptic vesicle with the presynaptic mem-
statistical model based on the assumption
brane. Subsequent evidence, based on other means
that the EPPs result from the independent
release of multiple MEPP-like quanta. The of measuring vesicle fusion, has left no doubt about
observed match, including the predicted the validity of this general interpretation of chemi-
0
0 0.4 0.8 number of failures, supports this interpreta- cal synaptic transmission. Recent work has identified
MEPP amplitude (mV) tion. (After Boyd and Martin, 1955.) structures within the presynaptic terminal that connect
vesicles to the plasma membrane and may be involved
in membrane contact and/or fusion (Figure 5.8C).
Release of Transmitters from
Synaptic Vesicles Local Recycling of Synaptic Vesicles
The discovery of the quantal release of packets of neu- The fusion of synaptic vesicles causes new membrane to
rotransmitter immediately raised the question of how be added to the plasma membrane of the presynaptic ter-
such quanta are formed and discharged into the synaptic minal, but the addition is not permanent. Although a bout
cleft. At about the time Katz and his colleagues were us- of exocytosis can dramatically increase the surface area of
ing physiological methods to discover quantal release of presynaptic terminals, this extra membrane is removed
neurotransmitter, electron microscopy revealed, for the within a few minutes. Heuser and Reese performed an-
first time, the presence of synaptic vesicles in presynap- other important set of experiments showing that the fused
tic terminals. Putting these two discoveries together, Katz vesicle membrane is actually retrieved and taken back into
and others proposed that synaptic vesicles loaded with the cytoplasm of the nerve terminal (a process called en-
transmitter are the source of the quanta. Subsequent bio- docytosis). The experiments, again carried out at the frog
chemical studies confirmed that synaptic vesicles are the neuromuscular junction, were based on filling the synaptic
repositories of transmitters. These studies have shown that cleft with horseradish peroxidase (HRP), an enzyme that
ACh is highly concentrated in the synaptic vesicles of mo- can be made to produce a dense reaction product that is vis-
tor neurons, where it is present at a concentration of about ible in an electron microscope. Under appropriate experi-
100 mM. Given the diameter of a small clear-core synaptic mental conditions, endocytosis could then be visualized by

Copyright
PURVES: © 2011 Sinauer
Neuroscience 5e Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
Figure: 05.07 in any form without express written permission from the publisher.
07/15/11
Synapti c T rans miss ion   87

(A) (C)
Unstimulated Stimulated

Synaptic vesicles

Vesicle fusing
with plasma
membrane

Fused vesicle
Ca2+ channels

FIGURE 5.8  Relationship between synaptic vesicle exocytosis and quan-


tal transmitter release. (A) A special electron microscopical technique called
freeze-fracture microscopy was used to visualize the fusion of synaptic vesicles in
presynaptic terminals of frog motor neurons. Left: Plasma membrane of an unstimu-
(B) lated presynaptic terminal. Right: Plasma membrane of a terminal stimulated by an
action potential; stimulation causes the appearance of dimple-like structures that
4-AP concentration: 10−3M
5000 represent the fusion of synaptic vesicles with the presynaptic membrane. The view
Number of vesicles fusing

in this image is as if looking down on the release sites from outside the presynaptic
terminal. (B) Comparison of the number of observed vesicle fusions to the number of
quanta released by a presynaptic action potential. Transmitter release was varied by
3000
using a drug (4-AP) that affects the duration of the presynaptic action potential, thus
10−4M changing the amount of calcium that enters during the action potential. The diago-
nal line is the 1:1 relationship expected if each vesicle that opened released a single
quantum of transmitter. (C) Fine structure of vesicle fusion sites of frog presynaptic
10−5M terminals. Synaptic vesicles are arranged in rows and are connected to each other
1000
and to the plasma membrane by a variety of proteinaceous structures (blue). The
purple structures in the presynaptic membrane, corresponding to the rows of par-
0
0 1000 3000 5000 ticles seen in (A), are thought to be Ca2+ channels. (A and B from Heuser et al., 1979;
Number of quanta released C after Harlow et al., 2001.)

the uptake of HRP into the nerve terminal (Figure 5.9). To from the reserve pool, docked at the presynaptic plasma
activate Neuroscience
PURVES: endocytosis,5ethe presynaptic terminal was stimu- membrane, and primed to participate in exocytosis once
Figure: 5.08 a train of action potentials, and the subsequent
lated with again. More recent experiments, employing a fluorescent
07/18/11
fate of the HRP was followed by electron microscopy. Im-
09/19/11 label rather than HRP, have determined the time course of
mediately following stimulation, the HRP was found within synaptic vesicle recycling. These studies indicate that the
special endocytotic organelles called coated vesicles (Figure entire vesicle cycle requires approximately 1 minute, with
5.9A,B). A few minutes later, however, the coated vesicles membrane budding during endocytosis requiring 10–20
had disappeared, and the HRP was found in a different seconds of this time. As can be seen from the 1 millisecond
organelle, the endosome (Figure 5.9C). Finally, within an delay in transmission following excitation of the presyn-
hour after stimulating the terminal, the HRP reaction prod- aptic terminal (see Figure 5.6B), membrane fusion during
uct appeared inside synaptic vesicles (Figure 5.9D). exocytosis is much more rapid than budding during endo-
These observations indicate that synaptic vesicle mem- cytosis. Thus, all of the recycling steps interspersed between
brane is recycled within the presynaptic terminal via the membrane budding and subsequent refusion of a vesicle
sequence summarized in Figure 5.9E. In this process, called are completed in less than a minute.
the synaptic vesicle cycle, the retrieved vesicular mem- The precursors to synaptic vesicles originally are pro-
brane passes through a number of intracellular compart- duced in the endoplasmic reticulum and Golgi apparatus
ments—such as coated vesicles and endosomes—and is in the neuronal cell body. Because of the long distance be-
eventually used to make new synaptic vesicles. After synap- tween the cell body and the presynaptic terminal in most
tic vesicles are re-formed, they are stored in a reserve pool neurons, transport of vesicles from the soma would not per-
within the cytoplasm until they need to participate again mit rapid replenishment of synaptic vesicles during con-
in neurotransmitter release. These vesicles are mobilized tinuous neural activity. Thus, local recycling is well suited to

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
88  C h a p t e r 5

(A) (B) (C) (D)


Wash away
extracellular HRP;
wait 5 minutes 1 hour later
Briefly
stimulate
presynaptic
terminal 3 Endosome
contains HRP
2 Coated pits and
coated vesicles
contain HRP

1 Synaptic 4 Synaptic vesicles


vesicles fuse contain HRP
Horseradish (E)
peroxidase (HRP)

FIGURE 5.9  Local recycling of synaptic vesicles in Endosome


presynaptic terminals. (A) Horseradish peroxidase (HRP)
introduced into the synaptic cleft is used to follow the fate
Endocytosis
of membrane retrieved from the presynaptic plasma mem-
brane. Stimulation of endocytosis by presynaptic action Budding
potentials causes HRP to be taken up into the presynaptic 1 min
terminals via a pathway that includes (B) coated vesicles and
(C) endosomes. (D) Eventually, the HRP is found in newly 10–20
formed synaptic vesicles. (E) Interpretation of the results sec
Budding
shown in A–D. Calcium-regulated fusion of vesicles with the Docking
presynaptic membrane is followed by endocytotic retrieval
of vesicular membrane via coated vesicles and endosomes, Fusion
and subsequent re-formation of new synaptic vesicles. (After Priming
Heuser and Reese, 1973.) Exocytosis
+ 1 msec
Ca2

the peculiar anatomy of neurons, giving nerve terminals the


means to provide a continual supply of synaptic vesicles. The explanation for this surprising finding was that cur-
rent was still flowing through Ca2+ channels, substituting
The Role of Calcium for the current ordinarily carried by the blocked Na+ chan-
in Transmitter Secretion nels. Subsequent voltage clamp experiments, performed by
Rodolfo Llinás and others at a giant presynaptic terminal of
As was apparent in the experiments of Katz and others the squid (Figure 5.10A), confirmed the presence of volt-
described in the preceding sections, lowering the concen- age-gated Ca2+ channels in the presynaptic terminal (Figure
tration of Ca2+ outside a presynaptic motor nerve termi- 5.10B). Such experiments showed that the amount of neu-
nal reduces the size of the EPP (compare Figures 5.6B and rotransmitter released is very sensitive to the exact amount
D). Moreover, measurement of the number of transmitter of Ca2+ that enters. Further, blockade of these Ca2+ channels
quanta released under such conditions shows that the rea- with drugs also inhibits transmitter release (Figure 5.10B,
son the EPP gets smaller is that lowering Ca2+ concentration right). These observations all confirm that the voltage-gated
decreases the number of vesicles that fuse with the plasma Ca2+ channels are directly involved in neurotransmission.
membrane of the terminal. An important insight into how Thus, presynaptic action potentials open voltage-gated Ca2+
Ca2+ regulates the fusion of synaptic vesicles was the dis- channels, with a resulting influx of Ca2+.
covery that presynaptic terminals have voltage-gated Ca2+ That Ca2+ entry into presynaptic terminals causes a rise
channels in their plasma membranes (see Chapter 4). in the concentration of Ca2+ within the terminal has been
The first indication of presynaptic Ca2+ channels was documented by microscopic imaging of terminals filled with
provided
PURVES: byNeuroscience
Katz and Ricardo
5e Miledi. They observed that Ca2+-sensitive fluorescent dyes (Figure 5.11A). The conse-
Figure: 05.09
presynaptic terminals treated with tetrodotoxin (which quences of the rise in presynaptic Ca2+ concentration for
07/18/11
blocks voltage-gated Na+ channels; see Chapter 3) could neurotransmitter release has been directly shown in two
still produce a peculiarly prolonged type of action potential. ways. First, microinjection of Ca2+ into presynaptic ter-

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   89

(A) Record FIGURE 5.10  Entry of Ca2+ through presynaptic voltage-gated


Postsynaptic calcium channels causes transmitter release. (A) Experimental setup
Presynaptic membrane using an extraordinarily large synapse in the squid. The voltage clamp
neuron potential method detects currents flowing across the presynaptic membrane when
Postsynaptic
neuron the membrane potential is depolarized. (B) Pharmacological agents that
block currents flowing through Na+ and K+ channels reveal a remaining
inward current flowing through Ca2+ channels. This influx of calcium triggers
transmitter secretion, as indicated by a change in the postsynaptic mem-
brane potential. Treatment of the same presynaptic terminal with cadmium,
Vpre Ipre
Voltage clamp a calcium channel blocker, eliminates both the presynaptic calcium current
and the postsynaptic response. (After Augustine and Eckert, 1984.)

(B) Control Ca2+ Channel blocker


0
Presynaptic –25
membrane
–50
potential (mV)
–75

Presynaptic 0
calcium
current
(µA/cm2) –200

0
Postsynaptic –25 FIGURE 5.11  Evidence that a rise in presynaptic
membrane
Ca2+ concentration triggers transmitter release from
potential (mV) –50
presynaptic terminals. (A) Fluorescence microscopy
–75 measurements of presynaptic Ca2+ concentration at the
–3 0 3 6 9 12 –3 0 3 6 9 12
Time (ms) squid giant synapse (see Figure 5.10A). A train of presyn-
aptic action potentials causes a rise in Ca2+ concentra-
tion, as revealed by a dye (called fura-2) that fluoresces
more strongly when the Ca2+ concentration increases.
(B) Microinjection of Ca2+ into a squid giant presynap-
PURVES: Neuroscience 5e tic terminal triggers transmitter release, measured as a
Figure: 05.10(A)
07/18/11 depolarization of the postsynaptic membrane potential.
09/19/11 5 0 µm (C) Microinjection of BAPTA, a Ca2+ chelator, into a squid
+
Ca2 giant presynaptic terminal prevents transmitter release.
(A from Smith et al., 1993; B after Miledi, 1973; C after
Adler et al., 1991.)
(C)
+
Control Inject Ca2 chelator
25
Postsynaptic membrane Presynaptic membrane
potential (mV)

−25

−50

(B) −75
Ca2+ injection
Postsynaptic membrane

25
potential (mV)

− 64
potential (mV)

−25

−50
− 65
−75
0 1 2 3 4 0 1 2 3 4 5 0 1 2 3 4 5
Time (s) Time (ms)

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.

PURVES: Neuroscience 5e
90  C h a p t e r 5

FIGURE 5.12  Differential release of neuropep- Small-molecule Neuropeptide


tide and small-molecule co-transmitters. Low- neurotransmitter in large dense-
in small clear- core vesicles
frequency stimulation preferentially raises the Ca2+ core vesicles
concentration close to the membrane, favoring the
release of transmitter from small clear-core vesicles
docked at presynaptic specializations. High-frequency Localized
stimulation leads to a more general increase in Ca2+, increase in Ca2+
causing the release of peptide neurotransmitters from concentration
large dense-core vesicles, as well as small-molecule
neurotransmitters from small clear-core vesicles.
Low-frequency
stimulation
Preferential release of small-
molecule neurotransmitter

More diffuse
+
increase in Ca2
concentration

minals triggers transmitter release in the absence of presyn-


aptic action potentials (Figure 5.11B). Second, presynaptic High-frequency
microinjection of calcium chelators (chemicals that bind stimulation Release of both
Ca and keep its concentration buffered at low levels) pre-
2+ types of transmitter
vents presynaptic action potentials from causing transmitter
secretion (Figure 5.11C). These results prove beyond any
doubt that a rise in presynaptic Ca2+ concentration is both Molecular Mechanisms
necessary and sufficient for neurotransmitter release.
PURVES:Thus,
of Synaptic Vesicle Cycling
Neuroscience 5e
as is the case for many other forms of neuronal Figure: 05.12
signaling
07/19/11
(see Chapter 7), Ca2+ serves as a second messenger during
07/26/11 Precisely how an increase in presynaptic Ca2+ concentration
transmitter release. goes on to trigger vesicle fusion and neurotransmitter re-
While Ca2+ is a universal trigger for transmitter release, lease is not understood. However, many important insights
not all transmitters are released with the same speed. For have come from molecular studies that have identified and
example, while secretion of ACh from motor neurons re- characterized the proteins found on synaptic vesicles (Fig-
quires only a fraction of a millisecond (see Figure 5.6), re- ure 5.13A) and their binding partners on the presynaptic
lease of neuropeptides requires high-frequency bursts of plasma membrane and cytoplasm. Most, if not all, of these
action potentials for many seconds. These differences in proteins act at one or more steps in the synaptic vesicle cy-
the rate of release probably arise from differences in the cle. Although a complete molecular picture of neurotrans-
spatial arrangement of vesicles relative to presynaptic Ca2+ mitter release is still lacking, the roles of several proteins in-
channels. This perhaps is most evident in cases where small volved in vesicle cycling have been deduced (Figure 5.13B).
molecules and peptides serve as co-transmitters (Figure Several lines of evidence indicate that the protein synap-
5.12). Whereas the small clear-core vesicles containing sin, which reversibly binds to synaptic vesicles, may keep
small-molecule transmitters are typically docked at the these vesicles tethered within the reserve pool by cross-
AU/ED:
plasma membrane in advance of Ca2+ entry, large We dense- link vesicles
showed neurtotransmitters to each
in vesicles other
per 5.09 to actin
and figures
and other filaments in the
in book.
core vesicles containing peptide transmitters are farther
Thanks, cytoskeleton. Mobilization of these reserve pool vesicles
away from the plasma membrane (see Figure 5.5D). Dragonfly
At lowMedia Groupis caused by phosphorylation of synapsin by proteins ki-
firing frequencies, the concentration of Ca may increase
2+
nases, most notably the Ca2+/calmodulin-dependent pro-
only locally at the presynaptic plasma membrane, in the vi- tein kinase, type II (CaMKII; see Chapter 7), which allows
cinity of open Ca2+ channels, limiting release to small-mole- synapsin to dissociate from the vesicles. Once vesicles are
cule transmitters from the docked small clear-core vesicles. free from their reserve pool tethers, they make their way to
Prolonged high-frequency stimulation increases the Ca2+ the plasma membrane and are then attached to this mem-
concentration throughout the presynaptic terminal, thereby brane by poorly understood docking reactions. A series of
inducing the slower release of neuropeptides. priming reactions then prepares the vesicular and plasma

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   91

(A) SNAP25 Vti1a Synaptobrevin


Synaptotagmin
FIGURE 5.13  Presynaptic proteins and
their roles in synaptic vesicle cycling.
V-ATPase (A) Model of the molecular organization of
a synaptic vesicle. The cytoplasmic surface
CIC3 of the vesicle membrane is densely covered
by proteins, only 70% of which are shown
Synaptophysin here. (B) The vesicle trafficking cycle shown
in Figure 5.9E is now known to be medi-
ated by a number of presynaptic proteins,
CSP SNAP29 including some of those shown in (A), with
different proteins participating in different
SV2 reactions. (A from Takamori et al., 2006.)

VAMP4
SCAMP
Syntaxin
Synapsin

(m)unc18
VGLUT

Rab3A

Trimeric Other
GTPase transporter

(B)

Transmitter loading Uncoating


Transmitter transporters Clathrin Endophilin
Proton pump Auxilin Synaptojanin
Budding Hsc-70

Budding
Mobilization
Synapsins Dynamin Clathrin
Actin Amphiphysin Actin
Endophilin Syndapin
Reserve pool Synaptojanin WASP
Synapsins
Actin

Docking Coating
GTP-binding Clathrin Epsin NSF
protein AP-2 Eps-15 SNAP
SNAREs? AP-180 Endophilin Syntaphilin
Synaptotagmins Stoned SNIP
Synaptobrevin
+
Ca2
Priming Fusion
SNAREs Complexin Syntaphilin Synaptotagmin 1
(m)unc13 Tomosyn Rab3a SNAREs
(m)unc18/nSec1 CAPS RIM
NSF SV2 Doc2
SNAPs Snapin

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
PURVES: Neuroscience 5e in any form without express written permission from the publisher.
AU/ED:
Figure: 05.13 OK to have this float within full width?
07/19/11 Thanks,
92  C h a p t e r 5

(A) (B) (1) Vesicle docks

Synaptic Synaptotagmin Synaptotagmin


vesicle
membrane
Synaptobrevin Vesicle
Ca2+ channel
Synaptobrevin
Ca2+
Syntaxin SNAP-25

(2) SNARE complexes form to pull


membranes together
SNAP-25

Syntaxin Presynaptic plasma membrane


+
(3) Entering Ca2 binds to synaptotagmin
FIGURE 5.14  Molecular mechanisms of exocytosis during neu-
rotransmitter release. (A) Structure of the SNARE complex. The vesicular
SNARE, synaptobrevin (blue), forms a helical complex with the plasma mem-
brane SNAREs syntaxin (red) and SNAP-25 (green). Also shown is the struc- Ca2+
ture of synaptotagmin, a vesicular Ca2+-binding protein, with bound Ca2+
indicated by spheres. (B) A model for Ca2+-triggered vesicle fusion. SNARE
proteins on the synaptic vesicle and plasma membranes form a complex (as
in A) that brings together the two membranes. Ca2+ then binds to synapto-
tagmin, causing the cytoplasmic region of this protein to catalyze membrane
fusion by binding to SNAREs and inserting into the plasma membrane. (4) Ca2+-bound synaptotagmin catalyzes
(A after Chapman, 2002.) membrane fusion by binding to SNAREs
and the plasma membrane

membranes for fusion. A large number of proteins are in-


volved in priming, including some proteins that are also in-
volved in other types of membrane fusion events common
to all cells (see Figure 5.13B). For example, two proteins
originally found to be important for the fusion of vesicles
with membranes of the Golgi apparatus, the ATPase NSF
(NEM-sensitive fusion protein) and SNAPs (soluble NSF-
attachment proteins), are also involved in priming synaptic
vesicles for fusion. These two proteins work by regulating the two membranes, thus bringing them into close appo-
the assembly of other proteins that are called SNAREs sition (Figure 5.14A). Such an arrangement is well suited
(SNAP receptors). Many of the other proteins involved in to promote the fusion of the two membranes, and several
priming—such as munc-13, nSec-1, complexin, snapin, lines of evidence suggest that this is what actually occurs.
syntaphilin, and tomosyn—also interact with the SNAREs. One important observation is that toxins that cleave the
One of the main purposes of priming seems to be to SNARE proteins block neurotransmitter release (Box 5B). In
organizeNeuroscience
PURVES: SNARE proteins
5e into the correct conformation for addition, putting SNARE proteins into artificial lipid mem-
membrane
Figure: 05.14 fusion. One of the SNARE proteins, synapto- branes and allowing these proteins to form complexes with
07/19/11
brevin, is in the membrane of synaptic vesicles, while two
09/23/11
each other causes the membranes to fuse.
other SNARE proteins called syntaxin and SNAP-25 are Because the SNARE proteins do not bind Ca2+, still
found primarily on the plasma membrane. These SNARE other molecules must be responsible for Ca2+ regulation
proteins can form a macromolecular complex that spans of neurotransmitter release. Several presynaptic proteins,

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   93

Box 5B  Diseases That Affect the Presynaptic Terminal


Defects in various steps in the exocyto- carcinoma, a form of lung cancer that A) or from a reduced supply of vesicles
sis and endocytosis of synaptic vesicles may somehow initiate the immune components from the motor neuron cell
have been shown to be at the root of a response to Ca2+ channels. Whatever body.
number of rare but debilitating neuro- the origin, the binding of antibodies to Still other patients suffering from
logical diseases. Ca2+ channels causes a reduction in Ca2+ familial infantile myasthenia appear to
channel currents. It is this antibody- have neuromuscular weakness that
Myasthenic syndromes induced defect in presynaptic Ca2+ entry arises from reductions in the size of
In myasthenic syndromes, abnormal that accounts for the muscle weakness individual quanta rather than the num-
transmission at neuromuscular synapses associated with LEMS. ber of quanta released. Motor nerve ter-
leads to weakness and fatigability of Congenital myasthenic syndromes are minals from these patients have synap-
skeletal muscles (see Box 7B). One of genetic disorders that, like LEMS, cause tic vesicles that are normal in number,
the best understood examples of these muscle weakness by affecting neuro- but smaller than normal in diameter.
disorders is Lambert-Eaton myasthenic muscular transmission. Some of these This finding suggests a different type
syndrome, or LEMS, an occasional com- syndromes affect the acetylcholinesterase of genetic lesion that somehow alters
plication in patients with certain kinds that degrades acetylcholine in the synap- formation of new synaptic vesicles
of cancers. Biopsies of muscle tissue tic cleft, whereas others arise from auto- following endocytosis, leading to less
removed from LEMS patients allow immune attack of acetylcholine receptors acetylcholine in each vesicle.
intracellular recordings identical to those (see Box 6B). However, a number of con-
in Figure 5.6. These recordings from genital myasthenic syndromes arise from Botulism and tetanus
affected tissues show that when a motor defects in acetylcholine release from the Impairment of synaptic transmitter
neuron is stimulated, the number of motor neuron terminal. Neuromuscular release also results from poisoning by
quanta contained in individual EPPs is synapses in some of these patients have anaerobic Clostridium bacteria. This
greatly reduced although the amplitude EPPs with reduced quantal content, genus of microorganisms produces
of spontaneous MEPPs is normal. Thus, a deficit that is especially prominent some of the most potent toxins known,
LEMS impairs evoked neurotransmit- when the synapse is activated repeat- including several botulinum toxins and
ter release but does not affect the size edly. Electron microscopy shows that tetanus toxin. Both botulism and teta-
of individual quanta. Several lines of presynaptic motor nerve terminals have nus are potentially deadly disorders.
evidence indicate that this reduction in a greatly reduced number of synaptic (Continued on next page)
neurotransmitter release is due to a loss vesicles. The defect in neu-
of voltage-gated Ca2+ channels in the rotransmitter release evidently
presynaptic terminal of motor neurons results from an inadequate Congenital myasthenic
number of synaptic vesicles syndromes result in
(see Figure A); perhaps most compelling impaired vesicle recycling
are anatomical studies that reveal a low- available for release during
ered density of Ca2+ channel proteins in sustained presynaptic activity.
the presynaptic plasma membrane. The origins of this shortage of Endosome
The loss of presynaptic Ca2+ channels synaptic vesicles is not clear,
in LEMS patients apparently arises from but could result either from an
an autoimmune disorder. Their blood impairment in endocytosis in
has a very high concentration of anti- the nerve terminal (see Figure
bodies that bind to Ca2+ channels, and it Budding
seems likely that these antibodies are the
(A) The presynaptic targets
primary cause of LEMS. For example, Budding
Latrotoxin
of several neurological triggers
removal of Ca2+ channel antibodies from
pathologies. Docking vesicle
the blood of LEMS patients by plasma fusion
Fusion
exchange reduces muscle weakness.
Priming
Similarly, immunosuppressant drugs
also can alleviate LEMS symptoms.
Perhaps most telling, injecting these Ca2+
antibodies into experimental animals
elicits muscle weakness and abnormal
neuromuscular transmission. Why the LEMS attacks
+
Cognitive disorders Botulinum and tetanus
immune system generates antibod- presynaptic Ca2 impair transsynaptic toxins affect SNARE
channels signaling proteins involved
ies against Ca2+ channels is not clear. in vesicle fusion
Most LEMS patients have small-cell

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.

PURVES: Neuroscience 5e
94  C h a p t e r 5

Box 5B (continued)
Botulism can occur by consuming The different actions of these toxins to two different types of presynaptic
food containing Clostridium bacteria, on synaptic transmission at excitatory proteins that may mediate its actions.
or by infection of wounds with the motor versus inhibitory synapses appar- One group of binding partners for
spores of these ubiquitous organisms. ently results from the fact that these α-latrotoxin is the neurexins, a group
In either case, the presence of the toxin toxins are taken up by different types of of integral membrane proteins found
can cause paralysis of peripheral neu- neurons: whereas the botulinum toxins in presynaptic terminals. Neurexins
romuscular synapses due to impaired are taken up by motor neurons, tetanus bind to synaptotagmin, the presynaptic
neurotransmitter release. This interfer- toxin is preferentially targeted to inter- Ca2+ sensor, and this interaction may
ence with neuromuscular transmission neurons. The differential uptake of tox- allow α-latrotoxin to bypass the usual
causes skeletal muscle weakness; in ins presumably arises from the presence Ca2+ requirement for triggering vesicle
extreme cases respiratory failure results of different types of toxin receptors on fusion. Another type of presynaptic
from paralysis of the diaphragm and the two types of neurons. protein that can bind to α-latrotoxin is
other muscles required for breathing. called CL1 (based on its previous names,
Botulinum toxins also block synapses Insights from α-latrotoxin Ca2+-independent receptor for latrotoxin
innervating the smooth muscles of sev- Latrodectism refers to the severe muscle and latrophilin-1). CL1 is a relative of
eral organs, giving rise to visceral motor pain and cramping experienced by the G-protein-coupled receptors that
dysfunction. patients bitten by the black widow mediate the actions of neurotransmitters
Tetanus typically results from the spider, Latrodectus. These symptoms and other extracellular chemical signals
contamination of puncture wounds arise from the presynaptic neurotoxin (see Chapter 7). Thus, the binding of
by Clostridium bacteria that produce α-latrotoxin, present in black widow α-latrotoxin to CL1 could activate an
tetanus toxin. In contrast to botulism, spider venom; this toxin causes a mas- intracellular signal transduction cas-
tetanus poisoning blocks the release sive discharge of neurotransmitter from cade involved in the Ca2+-independent
of inhibitory transmitters from inter- presynaptic terminals. Although neu- actions of α-latrotoxin. While more work
neurons in the spinal cord. This effect rotransmitter release ordinarily requires is needed to establish the definitive roles
causes a loss of synaptic inhibition on Ca2+, α-latrotoxin is capable of releas- of neurexins and CL1 in α-latrotoxin
spinal motor neurons, producing hyper- ing neurotransmitter even when Ca2+ is action, these two proteins are probably
excitation of skeletal muscle and tetanic absent from the extracellular medium. the basis for the toxin’s potent presynap-
contractions in affected muscles (hence Although it is not yet clear how the tic activity.
the name of the disease). toxin triggers Ca2+-independent exocy- In addition to their possible role
Although their clinical consequences tosis, we know that α-latrotoxin binds in latrodectism, neurexins have been
are dramatically different, clever and
patient biochemical work has shown
that clostridial toxins actually have a Synaptic vesicle
common mechanism of action: they are membrane
highly specific proteases that inhibit Synaptobrevin
neurotransmitter release by cleaving the
SNARE proteins involved in fusion of
synaptic vesicles with the presynaptic
plasma membrane (Figure B). Tetanus TeTX
toxin and botulinum toxin types B, D, BoTX-G BoTX-B BoTX-D BoTX-F
F, and G specifically cleave the vesicle
SNARE protein synaptobrevin. Other
botulinum toxins cleave syntaxin (type BoTX-A
C) and SNAP-25 (types A and E),
SNARE proteins found on the presyn- BoTX-C
aptic plasma membrane. Destruction of SNAP-25
these presynaptic proteins is the basis
for the inhibitory actions of clostridial BoTX-E
toxins on neurotransmitter release.

(B) Cleavage of SNARE proteins by clostridial toxins.


Indicated are the sites of proteolysis by tetanus toxin
(TeTX) and various types of botulinum toxin (BoTX). Syntaxin Presynaptic plasma membrane
(After Sutton et al., 1998.)

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   95

Box 5B (continued)
linked to a variety of cognitive disor- In summary, not only does research (2005) Internalization and mechanism
ders. Mutations in the neurexin gene into synaptic vesicle trafficking illumi- of action of clostridial toxins in neurons.
have been identified in a number of nate the causes of certain neurological Neurotoxicology 26: 761–767.
patients suffering from schizophrenia, and psychiatric disorders, but research Humeau, Y., F. Doussau, N. J. Grant and B.
a psychiatric disease causing delusions, into these diseases in turn has provided Poulain (2000) How botulinum and tetanus
hallucinations and loss of emotional tools—such as clostridial toxins and neurotoxins block neurotransmitter release.
Biochimie 82: 427–446.
expression (see Box 18D). Neurexins α-latrotoxin—that have proven valuable
are known to play an important role in elucidating the basic mechanisms of Maselli, R. A. (1998) Pathogenesis of human
in signaling across the synaptic cleft synaptic vesicle trafficking. botulism. Ann. N.Y. Acad. Sci. 841: 122–139.
by binding to a family of postsynaptic Silva, J. P., J. Suckling and Y. Ushkaryov
membrane proteins called neuroligins. (2009) Penelope’s web: Using α-latrotoxin
to untangle the mysteries of exocytosis. J.
Remarkably, mutations in neuroligins
References Neurochem. 111: 275–290.
also have been associated with autism,
a spectrum of psychiatric disorders Südhof, T. C. (2008) Neuroligins and
Dolly, J. O. and K. R. Aoki (2006) The struc-
neurexins link synaptic function to cognitive
characterized by impaired social inter- ture and mode of action of different botuli-
disease. Nature 455: 903–911.
action, communication problems, and num toxins. Eur. J Neurol. Suppl 4: 1–9.
Sutton, R. B., D. Fasshauer, R. Jahn and A. T.
other behavioral disorders. While a Engel, A. G. (1991) Review of evidence for
Brünger (1998) Crystal structure of a SNARE
direct connection between neurexins/ loss of motor nerve terminal calcium chan-
complex involved in synaptic exocytosis at 2.4
neuroligins and these psychiatric dis- nels in Lambert-Eaton myasthenic syn-
Å resolution. Nature 395: 347–353.
orders has not yet been established, it drome. Ann. N.Y. Acad. Sci. 635: 246–258.
Vincent, A. (2010) Autoimmune channelo-
seems likely that defects in these trans- Engel, A. G. (1994) Congenital myasthenic pathies: Well-established and emerging
synaptic signaling partners may serve syndromes. Neurol. Clin. 12: 401–437. immunotherapy-responsive diseases of the
as a central mechanism underlying a Grumelli, C., C. Verderio, D. Pozzi, O. peripheral and central nervous systems. J.
number of behavioral disorders. Rossetto, C. Montecucco and M. Matteoli Clin. Immunol. 30 (Suppl. 1): S97–S102.

including calmodulin, CAPS, and munc-13, are capable its three-legged appearance (Figure 5.15A). During endo-
of binding Ca2+. However, it appears that Ca2+ regulation cytosis, clathrin triskelia attach to the vesicular membrane
of neurotransmitter release is conferred by synaptotag- that is to be retrieved (Figure 5.15B). A number of adap-
min, a protein found in the membrane of synaptic vesicles tor proteins, such as AP-2 and AP180, connect clathrin to
(see Figure 5.14A). Synaptotagmin binds Ca2+ at concen- the proteins and lipids of this membrane. These adaptor
trations similar to those required to trigger vesicle fusion proteins, as well as other proteins such as amphiphysin,
within the presynaptic terminal, and this property allows epsin, and Eps-15, help to assemble individual triskelia
synaptotagmin to act as a Ca2+ sensor, signaling the eleva- into structures that resemble geodesic domes (see Figure
tion of Ca2+ within the terminal and thus triggering vesicle 5.15A). Such dome-like structures form coated pits that
fusion. In support of this idea, disruption of synaptotag- initiate membrane budding, increasing the curvature of
min in the presynaptic terminals of mice, fruit flies, squid, the budding membrane until it forms a coated vesicle-like
and other experimental animals impairs Ca2+-dependent structure. Another protein, called dynamin, causes the final
neurotransmitter release. In fact, deletion of only one of pinching-off of membrane that completes the production
the 19 synaptotagmin genes of mice is a lethal mutation, of coated vesicles. The clathrin coats then are removed by
causing the mice to die soon after birth. How Ca2+ binding an ATPase, Hsc70, with another protein, auxilin, serv-
to synaptotagmin leads to exocytosis is not yet clear. It is ing as a co-factor that recruits Hsc70 to the coated vesicle.
known that Ca2+ changes the chemical properties of synap- Other proteins, such as synaptojanin, are also important
totagmin, allowing it to insert into membranes and to bind for vesicle uncoating. Uncoated vesicles can then continue
to SNARE proteins. A plausible model is that the SNARE their journey through the recycling process, eventually be-
proteins bring the two membranes close together, and that coming refilled with neurotransmitter due to the actions
Ca2+-induced changes in synaptotagmin then produce the of neurotransmitter transporters in the vesicle membrane.
final fusion of these membranes (Figure 5.14B). These transporters exchange protons within the vesicle for
Still other proteins appear to be involved at the endo- neurotransmitter; the acidic interior of the vesicle is pro-
cytosis steps of the synaptic vesicle cycle (Figure 5.15). The duced by a proton pump that also is located in the vesicle
most important protein involved in endocytotic budding of membrane.
vesicles from the plasma membrane is clathrin. Clathrin In summary, a complex cascade of proteins, acting in
has a unique structure that is called a triskelion because of a defined temporal and spatial order, allows neurons to

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
96  C h a p t e r 5

(A) Clathrin FIGURE 5.15  Molecular mechanisms of endocytosis following neu-


triskelion rotransmitter release. (A) Individual clathrin triskelia (left) assemble together
to form membrane coats (right) involved in membrane budding during endocy-
tosis. (B) A model for membrane budding during endocytosis. Following addi-
tion of synaptic vesicle membrane during exocytosis, clathrin triskelia attach to
the vesicular membrane. Adaptor proteins, such as AP-2 and AP180, aid their
attachment. Polymerization of clathrin causes the membrane to curve, allowing
dynamin to pinch off the coated vesicle. Subsequent uncoating of the vesicle, by
Clathrin Hsc-70 and auxilin, yields a synaptic vesicle. (A after Marsh and McMahon, 1999.)
coat

(B)

Cytosol Actin

Clathrin

Hsc-70/auxilin

Outside cell Dynamin

1 Adaptor proteins 2 Clathrin triskelia 3 Dynamin ring 4 Coated vesicle 5 Hsc-70 and auxilin
connect clathrin assemble into coat, forms and pinches translocated uncoat vesicle
to vesicular curving membrane off membrane by actin filaments
membrane

secrete transmitters. Although the molecular mechanisms intervening metabolic steps. These receptors do not have
responsible for transmitter secretion are not entirely clear, ion channels as part of their structure; instead, they have an
most of the
PURVES: proteins involved
Neuroscience 5e in this process now have been intracellular domain that indirectly affects channels though
Figure: 05.15
identified. the activation of intermediate molecules called G-proteins
07/20/11
07/22/11
(Figure 5.16B). Neurotransmitter binding to these recep-
tors activates G-proteins, which then dissociate from the
Neurotransmitter Receptors receptor and interact directly with ion channels or bind to
The generation of postsynaptic electrical signals is also un- other effector proteins, such as enzymes, that make intra-
derstood in considerable depth. Such studies began in 1907, cellular messengers that open or close ion channels. Thus,
when the British physiologist John N. Langley introduced G-proteins can be thought of as transducers that couple
the concept of receptor molecules to explain the specific neurotransmitter binding to the regulation of postsynaptic
and potent actions of certain chemicals on muscle and ion channels. For this reason, metabotropic receptors are
nerve cells. We now know that neurotransmitter receptors also called G-protein-coupled receptors. The postsynap-
are proteins that are embedded in the plasma membrane of tic signaling events initiated by metabotropic receptors are
postsynaptic cells and have an extracellular neurotransmit- taken up in detail in Chapter 7.
ter binding site that detects the presence of neurotransmit- These two families of postsynaptic receptors give rise to
ters in the synaptic cleft. postsynaptic actions that range from less than a millisecond
There are two broad families of receptor proteins that to minutes, hours, or even days. Ionotropic receptors gen-
differ in their mechanism of transducing transmitter bind- erally mediate rapid postsynaptic effects. Examples are the
ing into postsynaptic responses. The receptors in one family EPP produced at neuromuscular synapses by ACh (see Fig-
contain a membrane-spanning domain that forms an ion ure 5.6B), as well as the postsynaptic responses produced
channel (Figure 5.16A). These receptors combine transmit- at certain glutamatergic synapses and GABAergic synapses
ter-binding and channel functions into a single molecular (see Figure 5.21B). In these cases, the PSPs arise within a
entity and thus are called ionotropic receptors (the Greek millisecond or two of an action potential invading the pre-
tropos means to move in response to a stimulus) or ligand- synaptic terminal and last for only a few tens of millisec-
gated ion channels. onds or less. In contrast, the activation of metabotropic re-
The second family of neurotransmitter receptors are ceptors typically produces much slower responses, ranging
metabotropic receptors, so called because the even- from hundreds of milliseconds to minutes or even longer.
tual movement of ions through a channel depends on The comparative slowness of metabotropic receptor actions

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   97

(A) Ligand-gated ion channels (B) G-protein-coupled receptors

Ions 1 Neurotransmitter
binds
1 Neurotransmitter
Neurotransmitter binds

Receptor
2 Channel
opens
Effector protein
Outside cell
5 Ions flow
across
membrane

γ
Inside cell β
α α
Intracellular
G-protein
messengers 4 Ion
channel
opens

3 Ions flow 2 G-protein is 3 G-protein subunits or Ions


across membrane activated intracellular messengers
modulate ion channels

FIGURE 5.16  Two different types of neurotransmitter


receptor. (A) Ligand-gated ion channels combine receptor and to ACh causes single-channel currents to flow for a few
channel functions in a single protein complex. (B) Metabotropic milliseconds (Figure 5.17A). This shows that ACh binding
receptors usually activate G-proteins, which modulate ion chan- to its receptors opens ligand-gated ion channels, much in
nels directly or indirectly through intracellular effector enzymes the way that changes in membrane potential open voltage-
and second messengers. gated ion channels (see Chapter 4).
The electrical actions of ACh are greatly multiplied when
an action potential in a presynaptic motor neuron causes
reflects the fact that multiple proteins need to bind to each the release of millions of molecules of ACh into the synaptic
other sequentially in order to produce the final physiologi- cleft. In this more physiological case, the transmitter mol-
cal response. Importantly, a given transmitter may activate ecules bind to many thousands of ACh receptors packed in
both ionotropic and metabotropic receptors to produce a dense array on the postsynaptic membrane, transiently
both fast and slow PSPs at the same synapse. opening a very large number of postsynaptic ion channels.
Although individual ACh receptors only open briefly (Fig-
ure 5.17B1), the opening of a large number of channels is
Postsynaptic Membrane synchronized by the brief duration during which ACh is
Permeability Changes during secreted from presynaptic terminals (Figure 5.17B2,3). The
Synaptic Transmission macroscopic current resulting from the summed opening of
many ion channels is called the end plate current, or EPC.
Just as studies of the neuromuscular synapse paved the way Because the current flowing during the EPC is normally
for understanding neurotransmitter release mechanisms, inward, it causes the postsynaptic membrane potential to
this peripheral synapse has been equally valuable for un- depolarize. This depolarizing change in potential is the EPP
derstanding the mechanisms that allow neurotransmitter (Figure 5.17C), which typically triggers a postsynaptic ac-
receptors to generate postsynaptic signals. The binding of tion potential by opening voltage-gated Na+ and K+ chan-
ACh to postsynaptic receptors opens ion channels in the nels (see Figure 5.6B).
muscle fiber membrane. This effect can be demonstrated The identity of the ions that flow during the EPC can
directly by using the patch clamp method (see Box 4A) to be determined via the same approaches used to identify
measure
PURVES: the minute postsynaptic
Neuroscience 5e currents that flow when the roles of Na+ and K+ fluxes in the currents underlying
twoFigure:
molecules
05.16 of individual ACh bind to receptors, as Erwin action potentials (see Chapter 3). Key to such an analysis
07/19/11
Neher and Bert Sakmann first did in 1976. Exposure of the is identifying the membrane potential at which no current
extracellular surface of a patch of postsynaptic membrane flows during transmitter action. When the potential of the

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
98  C h a p t e r 5

(A) Patch clamp measurement of single ACh receptor current (B) Currents produced by:
(1) SINGLE OPEN CHANNEL
Micropipette ACh release by
stimulating motor neuron
Number 0 0
Outside-out of open 1 2
membrane patch channels
2 4

Membranre current (pA)


ACh ACh receptor
(2) FEW OPEN CHANNELS
Na+
Number 0 0
2 µM Acetylcholine (ACh) of open
channels 10 20
0 Channel closed
I (pA)
2 Channel open (3) ALL CHANNELS OPEN

0 2 4 6 8 10 12 0 0
Number
Time (ms) of open 200,000
channels
FIGURE 5.17  Activation of ACh receptors at neuromus-
300,000 600,000
cular synapses. (A) Outside-out patch clamp measurement
of single ACh receptor currents from a patch of membrane –2 0 2 4 6 8 10 12 14
Time (ms)
removed from the postsynaptic muscle cell. When ACh is applied
to the extracellular surface of the membrane clamped at nega-
(C) Postsynaptic potential change (EPP) produced by EPC
tive voltages, the repeated brief opening of a single channel
−70
can be seen as downward deflections corresponding to inward
current (i.e., positive ions flowing into the cell). (B) Synchronized Membrane −80
opening of many ACh-activated channels at a synapse being potential
(mV) −90
voltage-clamped at negative voltages. (1) If a single channel is
examined during the release of ACh from the presynaptic termi- −100
nal, the channel opens transiently. (2) If a number of channels are –2 0 2 4 6 8 10 12 14
examined together, ACh release opens the channels almost syn- Time (ms)
chronously. (3) The opening of a very large number of postsynap-
tic channels produces a macroscopic EPC. (C) In a normal muscle ligand-gated channels. Thus, the value of the EPC is given
cell (i.e., not being voltage-clamped), the inward EPC depolarizes
by the relationship
the postsynaptic
PURVES: muscle
Neuroscience 5e cell, giving rise to an EPP. Typically, this
Figure: 05.17
depolarization generates an action potential (not shown). EPC = gACh(Vm – Erev)
07/15/11
07/28/11 where Erev is the reversal potential for the EPC. This rela-
tionship predicts that the EPC will be an inward current
postsynaptic muscle cell is controlled by the voltage clamp at potentials more negative than Erev because the electro-
method (Figure 5.18A), the magnitude of the membrane chemical driving force, Vm – Erev, is a negative number. Fur-
potential clearly affects the amplitude and polarity of EPCs ther, the EPC will become smaller at potentials approach-
(Figure 5.18B). Thus, when the postsynaptic membrane ing Erev because the driving force is reduced. At potentials
potential is made more negative than the resting poten- more positive than Erev, the EPC is outward because the
tial, the amplitude of the EPC becomes larger, whereas this driving force is reversed in direction (that is, positive). Be-
current is reduced when the membrane potential is made cause the channels opened by ACh are largely insensitive
more positive. At approximately 0 mV, no EPC is detected, to membrane voltage, gACh will depend only on the number
and at even more positive potentials, the current reverses of channels opened by ACh, which depends in turn on the
its polarity, becoming outward rather than inward (Figure concentration of ACh in the synaptic cleft. Thus, the mag-
5.18C). The potential where the EPC reverses, about 0 mV nitude and polarity of the postsynaptic membrane potential
in the case of the neuromuscular junction, is called the re- determines the direction and amplitude of the EPC solely
versal potential. by altering the driving force on ions flowing through the
As was the case for currents flowing through voltage- receptor channels opened by ACh.
gated ion channels (see Chapter 3), the magnitude of the When Vm is at the reversal potential, Vm – Erev is equal to 0
EPC at any membrane potential is given by the product and there is no net driving force on the ions that can perme-
of the ionic conductance activated by ACh (gACh) and the ate the receptor-activated channel. As a result, the identity
electrochemical driving force on the ions flowing through of the ions that flow during the EPC can be deduced by

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   99

observing how the reversal potential of the EPC compares these ions, because the reversal potential of the EPC is not
with the equilibrium potential for various ion species (Fig- near the equilibrium potential for any of them (see Figure
ure 5.18D). For example, if ACh were to open an ion chan- 5.18C). However, if these channels were permeable to both
nel permeable only to K+, then the reversal potential of the Na+ and K+, then the reversal potential of the EPC would be
EPC would be at the equilibrium potential for K+, which between +70 mV and –100 mV.
for a muscle cell is close to –100 mV. If the ACh-activated
channels were permeable only to Na+, then the reversal
FIGURE 5.18  The influence of the postsynaptic mem-
potential of the current would be approximately +70 mV, brane potential on end plate currents. (A) A postsynaptic
the Na+ equilibrium potential of muscle cells; if these chan- muscle fiber is voltage clamped using two electrodes, while the
nels were permeable only to Cl–, then the reversal potential presynaptic neuron is electrically stimulated to cause the release
would be approximately –50 mV. By this reasoning, ACh- of ACh from presynaptic terminals. This experimental arrange-
activated channels cannot be permeable to only one of ment allows the recording of macroscopic EPCs produced by
ACh. (B) Amplitude and time course of EPCs generated by stimu-
(A) Scheme for voltage clamping postsynaptic muscle fiber lating the presynaptic motor neuron while the postsynaptic cell
is voltage clamped at four different membrane potentials. (C) The
Stimulate
relationship between the peak amplitude of EPCs and postsyn-
Axon of aptic membrane potential is nearly linear, with a reversal poten-
presynaptic tial (the voltage at which the direction of the current changes
motor neuron Voltage clamp from inward to outward) close to 0 mV. Also indicated on this
amplifier graph are the equilibrium potentials of Na+, K+, and Cl– ions. (D)
The identity of the ions permeating postsynaptic receptors is
Current-passing
electrode revealed by the reversal potential (Erev). Activation of postsyn-
Postsynaptic
muscle fiber aptic channels permeable only to K+ (black) results in currents
Voltage-
measuring reversing at EK, near –100 mV, while activation of postsynaptic
electrode Na+ channels results in currents reversing at ENa, near +70 mV
Presynaptic
terminals (red). Cl–-selective currents reverse at ECl, near –50 mV (yellow).
(A–C after Takeuchi and Takeuchi, 1960.)

(B) Effect of membrane voltage on postsynaptic end plate currents (EPCs)

200 –110 mV –60 mV 0 mV +70 mV


Stimulate presynaptic axon Stimulate presynaptic axon
100
EPC (nA)

−100
Stimulate presynaptic axon Stimulate presynaptic axon
−200

−300

0 2 4 6 0 2 4 6 0 2 4 6 0 2 4 6
Time (ms)

(C) (D) Only K+-selective Only Cl–-selective


EK ECl ENa channel open channel open

300
200
EPC amplitude (nA)

Reversal ERev = EK
100 potential ERev = ENa

0
−100
ERev = ECl
−200 Only Na+-selective
channel open
−300
−110 −60 0 +70 −150 −100 −50 0 50 100
Postsynaptic membrane potential (mV) Membrane potential

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.

PURVES: Neuroscience 5e
100  C h a p t e r 5

(A) (B) FIGURE 5.19  Reversal potential of the


Lower external [Na+] shifts Higher external [K+] shifts end plate current changes when ion
reversal potential to left reversal potential to right
gradients change. (A) Lowering the external
Na+ concentration causes EPCs to reverse
200
EPC amplitude (nA)

at more negative potentials. (B) Raising the


100 external K+ concentration makes the reversal
potential more positive. (After Takeuchi and
0 Takeuchi, 1960.)
−100

−200

−300

−110 −60 0 +70 −110 −60 0 +70


Postsynaptic membrane potential (mV)

The fact that EPCs reverse at approximately 0 mV is there- postsynaptic resting membrane potential of –90 mV, the
fore consistent with the idea that ACh-activated ion chan- large inward EPC causes the postsynaptic membrane po-
PURVES: Neuroscience 5e
nels
Figure:are05.19
almost equally permeable to both Na+ and K+. This tential to become more depolarized (see Figure 5.20F).
was tested in 1960, by the Japanese husband and wife team
07/18/11 However, at 0 mV, the EPP reverses its polarity, and at more
07/26/11
of Akira and Noriko Takeuchi, by altering the extracellular positive potentials, the EPP is hyperpolarizing. Thus, the
09/19/11
concentration of these two ions. As predicted, the magnitude polarity and magnitude of the EPC depend on the electro-
AU/ED:
andused
We reversal potential
Red and of the
orange per EPC
figure was changed by altering the
5.18. chemical driving force, which in turn determines the polar-
concentration
Is this OK? gradient of each ion. Lowering the external ity and magnitude of the EPP. EPPs will depolarize when
Thanks,
Na+ concentration, which makes ENa more negative, pro- the membrane potential is more negative than Erev, and
Dragonfly Media Group
duces a negative shift in Erev (Figure 5.19A), whereas elevat- hyperpolarize when the membrane potential is more posi-
ing external K+ concentration, which makes EK more positive, tive than Erev. The general rule, then, is that the action of a
causes Erev to shift to a more positive potential (Figure 5.19B). transmitter drives the postsynaptic membrane potential toward
Such experiments confirm that the ACh-activated ion chan- Erev for the particular ion channels being activated.
nels are in fact permeable to both Na+ and K+. Although this discussion has focused on the neuromus-
Even though the channels opened by the binding of cular junction, similar mechanisms generate postsynaptic
ACh to its receptors are permeable to both Na+ and K+, responses at all chemical synapses. The general principle
at the resting membrane potential the EPC is generated is that transmitter binding to postsynaptic receptors pro-
primarily by Na+ influx (Figure 5.20). If the membrane po- duces a postsynaptic conductance change as ion channels
tential is kept at EK, the EPC arises entirely from an influx are opened (or sometimes closed). The postsynaptic con-
of Na+ because at this potential there is no driving force on ductance is increased if—as at the neuromuscular junc-
K+ (Figure 5.20A). At the usual muscle fiber resting mem- tion—channels are opened, and decreased if channels are
brane potential of –90 mV, there is a small driving force on closed. This conductance change typically generates an
K+, but a much greater one on Na+. Thus, during the EPC, electrical current, the postsynaptic current (PSC), which
much more Na+ flows into the muscle cell than K+ flows in turn changes the postsynaptic membrane potential to
out (Figure 5.20B); it is the net influx of positively charged produce a postsynaptic potential (PSP). As in the specific
Na+ that constitutes the inward current measured as the case of the EPP at the neuromuscular junction, PSPs are
EPC. At the reversal potential of about 0 mV, Na+ influx and depolarizing if their reversal potential is more positive than
K+ efflux are exactly balanced, so no current flows during the postsynaptic membrane potential and hyperpolarizing
the opening of channels by ACh binding (Figure 5.20C). At if their reversal potential is more negative.
potentials more positive than Erev the balance reverses; for The conductance changes and the PSPs that typically ac-
example, at ENa there is no influx of Na+ and a large efflux of company them are the ultimate outcome of most chemical
K+ because of the large driving force on K+ (Figure 5.20D). synaptic transmission, concluding a sequence of electrical
Even more positive potentials cause efflux of both Na+ and and chemical events that begins with the invasion of an ac-
K+ and produce an even larger outward EPC. tion potential into the terminals of a presynaptic neuron. In
Were it possible to measure the EPP at the same time many ways, the events that produce PSPs at synapses are
as the EPC (of course, the voltage clamp technique pre- similar to those that generate action potentials in axons; in
vents this by keeping membrane potential constant), the both cases, conductance changes produced by ion chan-
EPP would be seen to vary in parallel with the amplitude nels lead to ionic current flow that changes the membrane
and polarity of the EPC (Figures 5.20E,F). At the usual potential.

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   101
(A) Net ion fluxes EPCs EPPs
Postsynaptic Outside Na+
membrane cell ACh
potential Excitatory and Inhibitory
−100 mV
Postsynaptic Potentials
(EK)
PSPs ultimately alter the probability that
ACh- an action potential will be produced in
Inside activated the postsynaptic cell. At the neuromus-
cell channel
cular junction, synaptic action increases
(B) the probability that an action potential
Na+ will occur in the postsynaptic muscle cell;
indeed, the large amplitude of the EPP
ensures that an action potential always is
triggered. At many other synapses, PSPs
−90 mV
similarly increase the probability of firing
a postsynaptic action potential. However,
K+ still other synapses actually decrease the
probability that the postsynaptic cell will
(C) generate an action potential. PSPs are
Na+ called excitatory (or EPSPs) if they in-
crease the likelihood of a postsynaptic ac-
tion potential occurring, and inhibitory
0 mV (or IPSPs) if they decrease this likelihood.
(Erev) Given that most neurons receive inputs
from both excitatory and inhibitory syn-
K+ apses, it is important to understand more
precisely the mechanisms that determine
(D) whether a particular synapse excites or
inhibits its postsynaptic partner.
The principles of excitation just de-
scribed for the neuromuscular junction
+70 mV
(ENa)
are pertinent to all excitatory synapses.
The principles of postsynaptic inhibition
are much the same as for excitation, and
K+ they are also quite general. In both cases,
neurotransmitters binding to receptors
(E) (F)
open or close ion channels in the post-
synaptic cell. Whether a postsynaptic re-
Depolarizing sponse is an EPSP or an IPSP depends
EPC peak amplitude

EPP peak amplitude

on the type of channel that is coupled to


Outward
EK ENa EK ENa the receptor, and on the concentration of
(mV)
(nA)

permeant ions inside and outside the cell.


Inward
In fact, the only distinction between post-
Hyper− synaptic excitation and inhibition is the
polarizing reversal potential of the PSP in relation
−100 −90 0 +70 −100 −90 0 +70 to the threshold voltage for generating
Postsynaptic membrane potential Postsynaptic membrane potential action potentials in the postsynaptic cell.
Consider, for example, a neuronal syn-
FIGURE 5.20  Na+ and K+ movements during EPCs and EPPs. (A–D) Each of the apse that uses glutamate as the transmit-
postsynaptic potentials (Vpost) indicated at the left results in different relative fluxes of
ter. Many such synapses have receptors
net Na+ and K+ (ion fluxes). These ion fluxes determine the amplitude and polarity of the
EPCs, which in turn determine the EPPs. Note that at about 0 mV the Na+ flux is exactly
that, like the ACh receptors at neuromus-
balanced by an opposite K+ flux, resulting in no net current flow, and hence no change in cular synapses, open ion channels that
thePURVES:
membrane Neuroscience 5e EPCs are inward currents at potentials more negative than
potential. (E) are nonselectively permeable to cations
Erev Figure: 05.20 currents at potentials more positive than E . (F) EPPs depolarize the
and outward rev
(see Chapter 6). When these glutamate
06/20/11 receptors are activated, both Na+ and K+
postsynaptic cell at potentials more negative than Erev. At potentials more positive than
Erev, EPPs hyperpolarize the cell. flow across the postsynaptic membrane,

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
102  C h a p t e r 5

(A) (B) (C) (D)


ENa +50
Postsynaptic membrane potential (mV)

Action
potential
0
Erev
Erev > threshold =
excitatory
Threshold −40
Activate GABA synapse Erev
−50 EPSP
IPSP Erev < threshold =
Vrest −60 inhibitory
IPSP
−70
Erev
Activate glutamate
Activate GABA synapse
synapse
EK −110
0 1 2 3 4 0 1 2 3 4 0 1 2 3 4
Time (ms)

FIGURE 5.21  Reversal potentials and threshold poten-


yielding an Erev of approximately 0 mV for the resulting tials determine postsynaptic excitation and inhibition. (A)
postsynaptic current. If the resting potential of the postsyn- If the reversal potential for a PSP (0 mV) is more positive than the
PURVES:
aptic Neuroscience
neuron 5e the resulting EPSP will depolarize
is –60 mV, action potential threshold (–40 mV), the effect of a transmitter
Figure: 05.21
by bringing the postsynaptic membrane potential toward
07/15/11
is excitatory, and it generates EPSPs. (B) If the reversal potential
009/19/11
mV. For the hypothetical neuron shown in Figure 5.21A, for a PSP is more negative than the action potential threshold,
the action potential threshold voltage is –40 mV. Thus, a the transmitter is inhibitory and generates IPSPs. (C) IPSPs can
glutamate-induced EPSP will increase the probability that nonetheless depolarize the postsynaptic cell if their reversal
potential is between the resting potential and the action poten-
this neuron produces an action potential, defining the syn-
tial threshold. (D) The general rule of postsynaptic action is: If
apse as excitatory. the reversal potential is more positive than threshold, excitation
As an example of inhibitory postsynaptic action, con- results; inhibition occurs if the reversal potential is more negative
sider a neuronal synapse that uses GABA as its transmit- than threshold.
ter. At such synapses, the GABA receptors typically open
channels that are selectively permeable to Cl–, and the
action of GABA causes Cl– to flow across the postsynap-
tic membrane. Consider a case where ECl is –70 mV, as is mV, bringing the potential away from the action potential
the case for some neurons, so that the postsynaptic resting threshold. Thus, while EPSPs depolarize the postsynaptic
potential of –60 mV is less negative than ECl. The resulting cell, IPSPs can either hyperpolarize or depolarize; indeed,
positive electrochemical driving force (Vm – Erev) will cause an inhibitory conductance change may produce no poten-
negatively charged Cl– to flow into the cell and produce a tial change at all and still exert an inhibitory effect by mak-
hyperpolarizing IPSP (Figure 5.21B). This hyperpolarizing ing it more difficult for an EPSP to evoke an action potential
IPSP will take the postsynaptic membrane away from the in the postsynaptic cell.
action potential threshold of –40 mV, clearly inhibiting the Although the particulars of postsynaptic action can be
postsynaptic cell. complex, a simple rule distinguishes postsynaptic excitation
Surprisingly, inhibitory synapses need not produce hy- from inhibition: An EPSP has a reversal potential more pos-
perpolarizing IPSPs. For instance, if ECl were –50 mV in- itive than the action potential threshold, whereas an IPSP
stead of –70 mV, then the negative electrochemical driving has a reversal potential more negative than threshold (Fig-
force would cause Cl– to flow out of the cell and produce ure 5.21D). Intuitively, this rule can be understood by real-
a depolarizing IPSP (Figure 5.21C). However, the synapse izing that an EPSP will tend to depolarize the membrane
would still be inhibitory: given that the reversal potential potential so that it exceeds threshold, whereas an IPSP will
of the IPSP still is more negative than the action potential always act to keep the membrane potential more negative
threshold (–40 mV), the depolarizing IPSP would inhibit than the threshold potential.
because the postsynaptic membrane potential would be
kept more negative than the threshold for action potential Summation of Synaptic Potentials
initiation. Another way to think about this peculiar situation
is that if another excitatory input onto this neuron brought The PSPs produced at most synapses in the brain are much
the cell’s membrane potential to –41 mV—just below the smaller than those at the neuromuscular junction; indeed,
threshold for firing an action potential—the IPSP would EPSPs produced by individual excitatory synapses may be
then hyperpolarize the membrane potential toward –50 only a fraction of a millivolt and are usually well below the

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
Synapti c T rans miss ion   103

(A) FIGURE 5.22  Summation of postsynaptic potentials. (A)


A microelectrode records the postsynaptic potentials produced
by the activity of two excitatory synapses (E1 and E2) and an
inhibitory synapse (I). (B) Electrical responses to synaptic activa-
Excitatory E1 tion. Stimulating either excitatory synapse (E1 or E2) produces
Record
a subthreshold EPSP, whereas stimulating both synapses at the
Postsynaptic
membrane
same time (E1 + E2) produces a suprathreshold EPSP that evokes
Inhibitory I
potential a postsynaptic action potential (shown in blue). Activation of
the inhibitory synapse alone (I) results in a hyperpolarizing IPSP.
Excitatory E2 Summing this IPSP (dashed red line) with the EPSP (dashed yel-
low line) produced by one excitatory synapse (E1 + I) reduces the
Cell body amplitude of the EPSP (orange line), while summing it with the
suprathreshold EPSP produced by activating synapses E1 and
Axon E2 keeps the postsynaptic neuron below threshold, so that no
Dendrites action potential is evoked.

(B) Summed Summed Summed


EPSP EPSPs IPSP EPSP + IPSP EPSPs + IPSP
(Synapse (Synapses E1 + E2) (Synapse I) (Synapses (Synapses
E1 or E2) E1 + I) E1 + I + E2)
+20
Postsynaptic membrane
potential (mV)

−20

−40 Threshold

Vrest
−60

E1 or E2 E1 + E2 I E1 + I E1 + E2 + I

Time (ms)

threshold for generating postsynaptic action potentials. by an inhibitory synapse (I) can sum (algebraically speak-
How, then, can such synapses transmit information if their
PURVES: Neuroscience 5e
ing) with a subthreshold EPSP to reduce its amplitude (E1
PSPs
Figure:are subthreshold? The answer is that neurons in the
05.22 + I) or can sum with suprathreshold EPSPs to prevent the
central
07/20/11nervous system are typically innervated by thou- postsynaptic neuron from reaching threshold (E1 + I + E2).
09/23/11
sands
10/04/11
of synapses, and the PSPs produced by each active In short, the summation of EPSPs and IPSPs by a post-
synapse can sum together—in space and in time—to deter- synaptic neuron permits a neuron to integrate the electrical
mine the behavior of the postsynaptic neuron. information provided by all the inhibitory and excitatory
Consider the highly simplified case of a neuron that is synapses acting on it at any moment. Whether the sum of
innervated by two excitatory synapses, each generating a active synaptic inputs results in the production of an action
subthreshold EPSP, and an inhibitory synapse that pro- potential depends on the balance between excitation and
duces an IPSP (Figure 5.22A). While activation of either one inhibition. If the sum of all EPSPs and IPSPs results in a de-
of the excitatory synapses alone (E1 or E2 in Figure 5.22B) polarization of sufficient amplitude to raise the membrane
produces a subthreshold EPSP, activation of both excitatory potential above threshold, then the postsynaptic cell will
synapses at about the same time causes the two EPSPs to produce an action potential. Conversely, if inhibition pre-
sum together. If the sum of the two EPSPs (E1 + E2) de- vails, then the postsynaptic cell will remain silent. Normally,
polarizes the postsynaptic neuron sufficiently to reach the the balance between EPSPs and IPSPs changes continually
threshold potential, a postsynaptic action potential results. over time, depending on the number of excitatory and in-
Summation thus allows subthreshold EPSPs to influence hibitory synapses active at a given moment and the mag-
action potential production. Likewise, an IPSP generated nitude of the current at each active synapse. Summation is

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
104  C h a p t e r 5

Box 5C  The “Tripartite Synapse”


Up to this point, our discussion has close association with synapses (Figure These transient rises in intracellu-
considered signaling between presyn- A); a given synapse typically is no more lar calcium serve as second messenger
aptic neurons and their postsynaptic than a fraction of a micrometer away signals (see Chapter 7) that trigger a
targets to be a private dialogue between from a glial cell. Glia form exceedingly number of physiological responses in
these two cells. However, recent work fine processes that completely envelop glia. The most remarkable response is
suggests that this synaptic conversation synapses (Figure B), an intimate asso- the release of several molecules—such
may involve glial cells as well. ciation that raises the possibility of a as glutamate, GABA, and ATP—that are
As mentioned in Chapter 1, glial signaling role for glia at synapses. traditionally considered to be neuro­­-
cells support neurons in a number of The first support for such a role transmitters. Release of such “gliotrans­
ways. For example, it is well established came from the discovery that glia cells mitters” occurs both via the calcium-trig-
that glia regulate the extracellular envi- respond to application of neurotrans- gered exocytotic mechanisms employed
ronment by removing K+ that accumu- mitters. The list of neurotransmit- in presynaptic terminals of neurons (see
lates during action potential generation ters that elicit responses in glia now Figure 5.3), as well as via unconventional
and by removing neurotransmitters at includes acetylcholine, glutamate, release mechanisms such as permeation
the conclusion of synaptic transmission. GABA, and many others. These through certain ion channels.
Consistent with such roles, glia seem to responses are mediated by the same The ability both to respond to and
occupy virtually all of the non-neuronal sorts of neurotransmitter receptors that to release neurotransmitters potentially
volume of the brain. As a result of this are employed in synaptic signaling, makes glia participants in synaptic sig-
arrangement, glia are found in very most often the metabotropic receptors naling. Indeed, release of neurotransmit-
that are coupled to intracellular sig- ters from a variety of presynaptic termi-
naling cascades (see Figure 5.16B). In nals has been found to elicit responses
(A) Electron microscopy image of a synapse some cases, neurotransmitters produce in glial cells. Further, release of “glio-
between a presynaptic terminal (pre) and changes in the membrane potential transmitters” has been found to regulate
postsynaptic neuron (post), with a glial cell of glial cells. More often, these neu- transmission at numerous synapses
(astrocyte; astro) immediately adjacent to rotransmitters cause transient changes (Figure D). In some cases, glia regulate
the synapse. (B) Three-dimensional struc- in intracellular calcium concentration release of transmitters from presynaptic
ture of the contact between a glial cell within the glial cell. These intracellular terminals, while in other cases they alter
(blue) surrounding dendrites of four post- calcium signals often are observed to postsynaptic responsiveness.
synaptic neurons (different colors). (From trigger calcium waves that spread both This ability of glia to participate in
Witcher et al., 2007.) within a single glial cell and also propa- synaptic signaling has led to the con-
gate between glial cells (Figure C). cept of the tripartite synapse, a three-
(A) way junction involving the presynaptic
astro
(C)
pre
6s 12 s 18 s 24 s

post

200 µm
(B) (D)
(C) Application of glutamate (at arrow)
150 increases intracellular Ca2+ (white) in
(percent of control)

cultured glial cells. Examination of


Synaptic response

these cells at different times indicates


that the calcium signals spread as a
100
wave through neighboring glia. (From
Cornell-Bell et al., 1990.) (D) Transient
Raise Ca2+ in glial cell elevation of Ca2+ within a single glial
50 cell (arrow) enhances transmission at an
-60 0 60 120
excitatory synapse in the hippocampus.
Time (s) (From Perea and Araque, 2007.)

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
PURVES: Neuroscience 5e
Figure:BX05D.ABCD
Synapti c T rans miss ion   105

Box 5C (continued)
terminal, the postsynaptic process, and Fiacco, T. A., C. Agulhon and K. D. increases intracellular calcium in peri-
neighboring glial cells. While there is still McCarthy (2009) Sorting out astrocyte synaptic Schwann cells in situ. Neuron 8:
debate about the physiological signifi- physiology from pharmacology. Annu. Rev. 1069–1077.
cance of such neuron–glial interactions, Pharmacol. Toxicol. 49: 151–174. Lee, S. and 7 others (2010) Channel-
the newfound ability of glia to release Halassa, M. M. and P. G. Haydon (2010) mediated tonic GABA release from glia.
neurotransmitters, similar to presynaptic Integrated brain circuits: astrocytic networks Science 330: 790–796.
terminals, and to respond to neurotrans- modulate neuronal activity and behavior. Perea, G. and A. Araque (2007) Astrocytes
Annu. Rev. Physiol. 72: 335–355.  potentiate transmitter release at single hip-
mitters, similar to postsynaptic neurons,
promises to dramatically change our Hamilton, N. B. and D. Attwell (2010) Do pocampal synapses. Science 317: 1083–1086.
view of brain signaling mechanisms. astrocytes really exocytose neurotransmit- Perea, G., M. Navarrete and A. Araque
ters? Nat. Rev. Neurosci. 11: 227–238. (2009) Tripartite synapses: astrocytes process
References Haydon, P. G. and G. Carmignoto (2006) and control synaptic information. Trends
Astrocyte control of synaptic transmission Neurosci. 32: 421–431.
Cornell-Bell, A. H., S. M. Finkbeiner, M. and neurovascular coupling. Physiol. Rev. 86:
S. Cooper and S. J. Smith (1990) Glutamate Witcher, M. R., S. A. Kirov and K. M. Harris
1009–1031. (2007) Plasticity of perisynaptic astroglia
induces calcium waves in cultured astro-
cytes: Long-range glial signaling. Science Jahromi, B. S., R. Robitaille and M. P. during synaptogenesis in the mature rat hip-
247: 470–473. Charlton (1992) Transmitter release pocampus. Glia 55: 13–23.

therefore a neurotransmitter-induced tug-of-war between


all excitatory and inhibitory postsynaptic currents; the out-
come of the contest determines whether or not a postsyn- Neurotransmitter
aptic neuron fires an action potential and thereby becomes release
an active element in the neural circuits to which it belongs
(Figure 5.23). Recent work suggests that glial cells may also
contribute to synaptic signaling (Box 5C).
Receptor binding

Summary
Synapses communicate the information carried by action
potentials from one neuron to the next in neural circuits. Ion channels
The cellular mechanisms that underlie synaptic transmis- open or close
sion are closely related to the mechanisms that generate
other types of neuronal electrical signals, namely, ion flow
through membrane channels. In the case of electrical syn-
apses, these channels are gap junctions; direct but passive Conductance change
flow of current through the gap junctions is the basis for causes current flow
transmission. In the case of chemical synapses, channels
with smaller and more selective pores are activated by
the binding of neurotransmitters to postsynaptic recep-
Postsynaptic potential
tors following release of the neurotransmitters from the changes
presynaptic terminal. The large number of neurotransmit-
ters in the nervous system can be divided into two broad
classes: small-molecule transmitters and neuropeptides.
Postsynaptic cells
excited or inhibited
FIGURE 5.23  Events from neurotransmitter release to postsynaptic excita-
tion or inhibition. Neurotransmitter release at all presynaptic terminals on a cell
results in receptor binding, which causes the opening or closing of specific ion chan-
Summation determines
nels. The resulting conductance change causes current to flow, which may change the whether or not an
membrane potential. The postsynaptic cell sums (or integrates) all of the EPSPs and action potential occurs
IPSPs, resulting in moment-to-moment control of action potential generation.

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
PURVES: Neuroscience 5e
Figure: 05.23
106  C h a p t e r 5

Neurotransmitters are synthesized from defined precur- function of clathrin-coated vesicles. Annu. Rev. Cell. Dev. Biol.
sors by regulated enzymatic pathways, packaged into one 17: 517–568.
of several types of synaptic vesicles, and released into the Brunger, A. T. (2005) Structure and function of SNARE and
synaptic cleft in a Ca2+-dependent manner. Many synapses SNARE-interacting proteins. Quart. Rev. Biophys. 38: 1–47.
release more than one type of neurotransmitter, and mul- Carlsson, A. (1987) Perspectives on the discovery of central
tiple transmitters can even be packaged within the same monoaminergic neurotransmission. Annu. Rev. Neurosci. 10:
19–40.
synaptic vesicle. Transmitter agents are released presynap-
Chapman, E. R. (2002) Synaptotagmin: A Ca2+ sensor that trig-
tically in units or quanta, reflecting their storage within syn- gers exocytosis? Nat. Rev. Mol. Cell Biol. 3: 498–508.
aptic vesicles. Vesicles discharge their contents into the syn-
Connors, B. W. and M. A. Long (2004) Electrical synapses in
aptic cleft when the presynaptic depolarization generated the mammalian brain. Annu. Rev. Neurosci. 27: 393–418.
by the invasion of an action potential opens voltage-gated Emson, P. C. (1979) Peptides as neurotransmitter candidates in
calcium channels, allowing Ca2+ to enter the presynaptic the CNS. Prog. Neurobiol. 13: 61–116.
terminal. How calcium triggers neurotransmitter release Hestrin, S. and M. Galarreta (2005) Electrical synapses define
is not yet established, but synaptotagmin, SNAREs, and networks of neocortical GABAergic neurons. Trends Neurosci.
a number of other proteins found within the presynaptic 28: 304–309.
terminal are clearly involved. Postsynaptic receptors are a Jackson, M. B. and E. R. Chapman (2006) Fusion pores and
diverse group of proteins that transduce binding of neu- fusion machines in Ca2+-triggered exocytosis. Annu. Rev.
rotransmitters into electrical signals by opening or closing Biophys. Biomol. Struct. 35: 135–160.
postsynaptic ion channels. Two broadly different families Marsh, M. and H. T. McMahon (1999) The structural era of
of neurotransmitter receptors have evolved to carry out the endocytosis. Science 285: 215–220.
postsynaptic signaling actions of neurotransmitters. The Pierce, K. L., R. T. Premont and R. J. Lefkowitz (2002) Seven-
membrane receptors. Nature Rev. Mol. Cell Biol. 3: 639–650.
postsynaptic currents produced by the synchronous open-
ing or closing of ion channels changes the conductance Rizzoli, S. O. and W. J. Betz (2005) Synaptic vesicle pools.
Nature Rev. Neurosci. 6: 57–69.
of the postsynaptic cell, thus increasing or decreasing its
Royle, S. J. and L. Lagnado (2010) Clathrin-mediated endocy-
excitability. Conductance changes that increase the prob- tosis at the synaptic terminal: Bridging the gap between physi-
ability of firing an action potential are excitatory, whereas ology and molecules. Traffic 11: 1489–1497.
those that decrease the probability of generating an action Südhof, T. and J. E. Rothman (2009) Membrane fusion:
potential are inhibitory. Because postsynaptic neurons are Grappling with SNARE and SM proteins. Science 323: 474–477.
usually innervated by many different inputs, the integrated
effect of the conductance changes underlying all EPSPs and
IPSPs produced in a postsynaptic cell at any moment deter- Important Original Papers
mines whether or not the cell fires an action potential. The Adler, E., M. Adler, G. J. Augustine, M. P. Charlton and S.
postsynaptic effects of neurotransmitters are terminated by N. Duffy (1991) Alien intracellular calcium chelators attenuate
neurotransmitter release at the squid giant synapse. J. Neurosci.
the degradation of the transmitter in the synaptic cleft, by 11: 1496–1507.
transport of the transmitter back into cells, or by diffusion
Augustine, G. J. and R. Eckert (1984) Divalent cations differen-
out of the synaptic cleft. The response elicited at a given tially support transmitter release at the squid giant synapse. J.
synapse depends on the type of neurotransmitter released Physiol. (Lond.) 346: 257–271.
and the postsynaptic complement of receptors and associ- Beierlein, M., J. R. Gibson and B. W. Connors (2000) A network
ated channels. of electrically coupled interneurons drives synchronized inhibi-
tion in neocortex. Nature Neurosci. 3: 904–910.
Boyd, I. A. and A. R. Martin (1955) The end-plate potential in
Go to the NEUROSCIENCE Companion Website mammalian muscle. J. Physiol. (Lond.) 132: 74–91.
at sites.sinauer.com/neuroscience5e for Animations, Curtis, D. R., J. W. Phillis and J. C. Watkins (1959) Chemical
Flashcards, Online Quizzes, and other study aids. excitation of spinal neurons. Nature 183: 611–612.
Dale, H. H., W. Feldberg and M. Vogt (1936) Release of ace-
tylcholine at voluntary motor nerve endings. J. Physiol. 86:
353–380.
Additional Reading del Castillo, J. and B. Katz (1954) Quantal components of the

Go to the NEUROSCIENCE Companion Website end plate potential. J. Physiol. (Lond.) 124: 560–573.
Reviews Fatt, P. and B. Katz (1951) An analysis of the end plate poten-
Aat sites.sinauer.com/neuroscience5e
ugustine for Animations,
, G. J. and H. Kasai (2006) Bernard Katz, quantal tial recorded with an intracellular electrode. J. Physiol. (Lond.)
Flashcards,
transmitter release, Online
and theQuizzes and other
foundations study aids.
of presynaptic physiol- 115: 320–370.
ogy. J. Physiol. (Lond.) 578: 623–625.
Fatt, P. and B. Katz (1952) Spontaneous subthreshold activity
Brodsky, F. M., C. Y. Chen, C. Knuehl, M. C. Towler and D. at motor nerve endings. J. Physiol. (Lond.) 117: 109–128.
E. Wakeham (2001) Biological basket weaving: Formation and

GoCopyright © 2011 Sinauer Associates,


to the NEUROSCIENCE Companion Inc. Website
This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.
at sites.sinauer.com/neuroscience5e for Animations,
Flashcards, Online Quizzes and other study aids.
Synapti c T rans miss ion   107

Furshpan, E. J. and D. D. Potter (1959) Transmission at the Smith, S. J., J. Buchanan, L. R. Osses, M. P. Charlton and G. J.
giant motor synapses of the crayfish. J. Physiol. (Lond.) 145: Augustine (1993) The spatial distribution of calcium signals in
289–325. squid presynaptic terminals. J. Physiol. (Lond.) 472: 573–593.
Geppert, M. and 6 others (1994) Synaptotagmin I: A major Sutton, R. B., D. Fasshauer, R. Jahn and A. T. Brünger (1998)
Ca2+ sensor for transmitter release at a central synapse. Cell 79: Crystal structure of a SNARE complex involved in synaptic exo-
717–727. cytosis at 2.4 Å resolution. Nature 395: 347–353.
Harris, B. A., J. D. Robishaw, S. M. Mumby and A. G. Gilman Takamori, S. and 21 others (2006) Molecular anatomy of a traf-
(1985) Molecular cloning of complementary DNA for the alpha ficking organelle. Cell 127: 831–846.
subunit of the G protein that stimulates adenylate cyclase. Takeuchi, A. and N. Takeuchi (1960) On the permeability of
Science 229: 1274–1277. end-plate membrane during the action of transmitter. J. Physiol.
Heuser, J. E., T. S. Reese, M. J. Dennis, Y. Jan, L. Jan and L. (Lond.) 154: 52–67.
Evans (1979) Synaptic vesicle exocytosis captured by quick Wickman, K. and 7 others (1994) Recombinant Gβγ activates the
freezing and correlated with quantal transmitter release. J. Cell muscarinic-gated atrial potassium channel IKACh. Nature 368:
Biol. 81: 275–300. 255–257.
Heuser, J. E. and T. S. Reese (1973) Evidence for recycling of
synaptic vesicle membrane during transmitter release at the Books
frog neuromuscular junction. J. Cell Biol. 57: 315–344. Cooper, J. R., F. E. Bloom and R. H. Roth (2002) The Biochemical
Hökfelt, T., O. Johansson, A. Ljungdahl, J. M. Lundberg and M. Basis of Neuropharmacology. New York: Oxford University Press.
Schultzberg (1980) Peptidergic neurons. Nature 284: 515–521. Hall, Z. (1992) An Introduction to Molecular Neurobiology.
Jonas, P., J. Bischofberger and J. Sandkuhler (1998) Co-release Sunderland, MA: Sinauer Associates.
of two fast neurotransmitters at a central synapse. Science 281: Katz, B. (1969) The Release of Neural Transmitter Substances.
419–424. Liverpool: Liverpool University Press.
Loewi, O. (1921) Über humorale Übertragbarkeit der Llinás, R. R. (1999) The Squid Giant Synapse: A Model for
Herznerven-wirkung. Pflügers Arch. 189: 239–242. Chemical Synaptic Transmission. Oxford: Oxford University
Miledi, R. (1973) Transmitter release induced by injection of Press.
calcium ions into nerve terminals. Proc. R. Soc. Lond. B 183: Nicholls, D. G. (1994) Proteins, Transmitters, and Synapses.
421–425. Oxford: Blackwell.
Neher, E. and B. Sakmann (1976) Single-channel currents Peters, A., S. L. Palay and H. deF. Webster (1991) The Fine
recorded from membrane of denervated frog muscle fibres. Structure of the Nervous System: Neurons and their Supporting
Nature 260: 799–802. Cells, 3rd Ed. Oxford: Oxford University Press.

Copyright © 2011 Sinauer Associates, Inc. This material cannot be copied, reproduced, manufactured or disseminated
in any form without express written permission from the publisher.

You might also like