You are on page 1of 11

Clinical Neurophysiology 112 (2001) 1575±1585

www.elsevier.com/locate/clinph
Review

Excitability of human axons


David Burke a,*, Matthew C. Kiernan a,b, Hugh Bostock b
a
Department of Neurology, Prince of Wales Hospital and Prince of Wales Medical Research Institute, University of New South Wales,
Sydney, N.S.W., Australia
b
Sobell Department of Neurophysiology, Institute of Neurology, London, UK
Accepted 15 May 2001

Abstract
The excitability of human axons can be studied reliably using the technique of threshold tracking, which allows the strength of a test
stimulus to be adjusted by computer to activate a de®ned fraction of the maximal nerve or muscle action potential. The stimulus current that
just evokes the target response is considered the `threshold' for that response. More useful than the resting threshold are other indices of
axonal excitability derived from pairs of threshold measurements, such as refractoriness, supernormality, strength-duration time constant and
`threshold electrotonus' (i.e. the changes in threshold produced by long-lasting depolarizing or hyperpolarizing current pulses). Each of these
measurements depends on membrane potential and on other biophysical properties of the axons. Together they can provide new information
about the pathophysiology underlying abnormalities in excitability in neuropathy. q 2001 Elsevier Science Ireland Ltd. All rights reserved.
Keywords: Axonal excitability; Recovery cycle; Threshold electrotonus; Depolarization; Hyperpolarization

1. Introduction Kiernan et al., 2000), and Table 2 lists some of the neuro-
pathic disturbances that have been so studied. This paper
When axons are diseased or damaged, disturbed axonal reviews some of the determinants of the excitability of large
excitability may result in the inability to maintain conduc- myelinated axons and the mechanisms underlying the excit-
tion of a meaningful impulse train (Bostock and Grafe, ability properties that can be studied in vivo in human
1985; Burke et al., 1997; Inglis et al., 1998; Cappelen- subjects.
Smith et al., 2000; Kaji et al., 2000). Alternatively ectopic
activity may develop at a focus of hyperexcitability leading
to symptoms of paraesthesiae, or pain or fasciculation 2. Threshold tracking methods
(Lance et al., 1979; Ochoa and TorebjoÈrk, 1980; NystroÈm
and Hagbarth 1981; Culp and Ochoa, 1980; Nordin et al., The electrical excitability of an axon is de®ned by the
1984; Waxman et al., 1999; Mogyoros et al., 2000). The threshold current that just excites it. Threshold determina-
abnormalities of axonal excitability that underlie conduc- tion is a trial-and-error process, and best achieved using a
tion block and ectopic impulse activity are not adequately computer to control the output of a current source, depend-
explored by routine nerve conduction studies. In routine ing on the response of the axon or group of axons stimu-
diagnostic studies, only latency or conduction velocity can lated. The interested reader is referred to a recent review of
be measured accurately but, while such measurements may `threshold tracking' for a detailed discussion of the techni-
be very useful in de®ning pathology, they provide little que (Bostock et al., 1998). For single axons, `threshold' is
insight into underlying disease mechanisms because a estimated as the stimulus current that activates the axon
number of morphological and functional changes can 50% of the time. However, in clinical practice, tracking
prolong latency (Table 1). Increasingly the technique of the threshold of a compound sensory or motor action poten-
`threshold tracking' is being used in research and clinical tial is usually more rewarding than tracking the threshold for
studies on large myelinated axons (Bostock et al., 1998; a single axon. With compound potentials, the computer is
set to estimate the stimulus current necessary to produce a
target potential that is a speci®c % of maximum.
* Corresponding author. Prince of Wales Medical Research Institute,
Barker Street, Randwick, N.S.W. 2031, Australia. Tel.: 161-2-9382-
Commonly, the target size is set at 30, 40 or 50% of the
2671; fax: 161-2-9382-2724. maximal compound action potential because these sizes fall
E-mail address: d.burke@unsw.edu.au (D. Burke). on the steeply rising phase of the stimulus/response curve
1388-2457/01/$ - see front matter q 2001 Elsevier Science Ireland Ltd. All rights reserved. CLINPH 2001024
PII: S 1388-245 7(01)00595-8
1576 D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585

Table 1 duration conditioned by a supramaximal stimulus using a 7


Causes of conduction slowing ms interstimulus interval. This particular set of stimuli
Morphological variations or changes would allow several different excitability parameters
Axonal tapering; axonal attenuation at sites of constriction; axonal described in this review (threshold, refractoriness at 2 ms,
regrowth supernormality at 7 ms, strength-duration time constant) to
Demyelination be followed at much the same time. Clearly, the time taken
Nodal displacement and intussusception
before a particular threshold estimate is updated will be
Remyelination with short internodes
longer the greater the number of stimuli and the longer
Functional (changes in axonal excitability) the interval between successive stimuli. In the above exam-
Cooling ple, each excitability parameter would be updated every 5 s
Axonal hyperpolarization if the interval between successive stimuli was 1 s.
Axonal depolarization
For recording `threshold electrotonus' (as in Figs. 2 and
4A) repeated comparisons are made between the resting
threshold and the threshold at different times after the start
of long-lasting subthreshold depolarizing and hyperpolariz-
(see Kiernan et al., 2000, 2001d). If a manoeuvre changes
ing currents. The accuracy of each threshold estimate (i.e.
the size of the maximal potential, it is necessary to measure
for each value of the delay between the start of the condi-
the maximal response regularly and to change the size of the
tioning current and the test stimulus) depends on the number
target potential tracked by the computer accordingly. This
of stimuli delivered, and on the change in threshold since the
can be achieved by appropriate threshold-tracking software
last threshold estimate. The QTRAC software allows the
(e.g. QTRAC, qInstitute of Neurology, London, UK).
conditioning-test delay to be advanced only when a speci-
As mentioned later, the absolute value of the threshold
®ed number of stimuli provide acceptable threshold esti-
current usually provides little information, because it is vari-
mates. The recording of recovery cycles (as in Figs. 3 and
able within a subject and between subjects. More informa-
4B) is similar, except that the conditioning stimuli are brief
tion is provided by excitability parameters obtained by
and supramaximal, instead of long-lasting and subthreshold.
comparing two threshold estimates (e.g. strength-duration
To allow accurate measurement of the responses to the test
time constant, which can be estimated from the thresholds
stimuli, the response to the supramaximal conditioning
to 1 ms and 0.1 ms stimuli). The computer can control the
stimulus alone is subtracted from the response to each
delivery of different stimuli (or stimulus combinations) in a
conditioning 1 test pair before measurement. The threshold
regularly recurring sequence, with successive stimuli deliv-
electrotonus and recovery cycle recordings in Fig. 4 each
ered at intervals of about 1 s. For example, different stimuli
took between 3 and 3.5 min, with stimuli presented every
might be (i) a supramaximal stimulus, (ii) an unconditioned
0.8 s.
test stimulus of 0.1 ms duration, (iii) an unconditioned test
stimulus of 1.0 ms duration, (iv) a test stimulus of 0.1 ms
duration conditioned by a supramaximal stimulus using a 2
ms interstimulus interval, and (v) a test stimulus of 0.1 ms 3. Axonal excitability depends on nodal and internodal
ion channels
Table 2
Axonal excitability studies in neuropathic disorders The action potential of human myelinated axons can be
modelled successfully using only the properties of transient
Disorder References Na 1 channels (Schwarz et al., 1995). Other channels are
Auto-immune neuropathy
activated during and after the action potential. Some contri-
Acquired neuromyotonia Maddison et al., 1999; Kiernan bute to resting membrane potential and thereby determine
et al., 2001c the threshold for action potential generation. Ion channels
Diabetic polyneuropathy Weigl et al., 1989; Strupp et al., are not evenly distributed along a myelinated axon (Fig. 1).
1990; Horn et al., 1996 The density of Na 1 channels at the node of Ranvier is ,30
Demyelinating polyneuropathies
Multifocal motor neuropathy Kaji et al., 2000
times the density on the internodal membrane. The interno-
Chronic in¯ammatory Cappelen-Smith et al., 2000 dal density is too low for action potential propagation and,
demyelinating polyneuropathy in demyelinated axons, the action potential is generated only
Toxic neuropathies at the node unless the density of sodium channels on the
Taxol/cisplatin Hanauske et al., 1995; Schilling internodal membrane increases.
et al., 1997
Mononeuropathies
Recent evidence suggests that, although 6 types of Na 1
Carpal tunnel syndrome Mogyoros et al., 1997a; Kiernan channel are expressed in the peripheral nervous system
et al., 1999 (Goldin, 1999; Goldin et al., 2000; Waxman et al., 1999),
Neuronopathy only one (Nav1.6) is found at nodes of Ranvier (Caldwell et
Amyotrophic lateral sclerosis Bostock et al., 1995; Horn et al., al., 2000). Nevertheless two functionally distinct types of
1996; Mogyoros et al., 1998a,b
sodium current can be distinguished. The classical `transi-
D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585 1577

Fig. 1. Distribution of some conductances. Different channels are not distributed evenly along the membrane of myelinated ®bres, as described further in the
text.

ent' Na 1 current (INat in Fig. 1) is activated rapidly by Fig. 1) have a density at the node 25 times that at the inter-
membrane depolarization and then inactivates, so that node, but their kinetics are too slow to allow them to affect
further Na 1 ions cannot pass through the channel no matter the action potential directly. They help determine resting
how much the membrane is depolarized. About 98% of the membrane potential, contribute to accommodation to depo-
sodium current behaves in this way. `Persistent' Na 1 current larizing stimuli, and are largely responsible for the late
(INap) is activated equally rapidly but at membrane potentials subnormal period and for the hyperpolarization that follows
that are ,10±20 mV more negative. At the most negative a brief train of impulses (see below).
potentials over which the current activates, inactivation is There are channels on the internode that are activated
minimal, giving rise to a persistent inward leak of sodium slowly by hyperpolarization, pass both Na 1 and K 1 ions,
ions at the resting potential, a potentially destabilizing prop- and produce accommodation to the hyperpolarization
erty. Both `persistent' and `transient' components of the (hence the term `inward recti®cation'). The conductance is
Na 1 current have been recorded in rat dorsal root ganglia now usually referred to as IH, the `hyperpolarization-acti-
cells (Baker and Bostock, 1997, 1998; Kiernan et al., vated cation conductance' (Pape, 1996), and its biological
2001a), and excitability studies indicate that both are role is probably to limit the hyperpolarization that occurs
present in human peripheral nerve (Bostock and Rothwell, when axons conduct trains of impulses.
1997).
There are many types of K 1 channel on axons (Vogel and
Schwarz, 1995; Reid et al., 1999), but it is convenient for 4. The internode is not isolated and its properties set the
clinical purposes to restrict discussion to two groups that are resting membrane potential
dependent on membrane potential, those with fast kinetics
and those with slow kinetics. There are few fast K 1 channels Changes in potential of the node spread to the internode,
(K 1f in Fig. 1) at the human node of Ranvier such that, when but slowly because of the resistance of the myelin sheath
modelling the action potential, K 1 channels can be ignored and the large capacitance of the internodal axolemma. This
(Schwarz et al., 1995). In mammalian axons repolarization results in slow activation/deactivation of voltage-dependent
is achieved by inactivation of Na 1 channels and current leak channels on the internodal membrane. Although Na 1 chan-
to the internode (Ritchie, 1995). Fast K 1 channels are nel density is insuf®cient for the internodal membrane to
located in a tight band in the paranodal region where they generate an action potential, the changes in resistance of
contribute to the resistance of the internodal membrane and the internodal membrane and in the current stored on it
limit the depolarizing afterpotential responsible for the will affect the behaviour of the node (for review see
supernormal period (see later). Slow K 1s channels (Ks in Baker, 2000). Approximately 99.9% of axonal membrane
1578 D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585

is internodal so that, despite the lower channel densities, de¯ection and a decrease in excitability a downward de¯ec-
there are many more channels on the internode than on tion. This convention was adopted so that changes in thresh-
the node. old electrotonus would resemble the underlying changes in
Resting membrane potential is determined by those chan- membrane potential (Bostock and Baker, 1988).
nels that are open at or near rest (both voltage-dependent
and voltage-independent) and by the activity of the Na 1/K 1 5.1. There is accommodation to long-lasting depolarizing
pump, and this implies that it is largely determined by the current pulses
properties of the internodal membrane. The major contribu-
tors are probably slow and fast K 1 channels, persistent Na 1 In response to depolarizing current pulses, there is an
channels, and the Na 1/K 1 pump. initial fast phase of threshold electrotonus that is almost
instantaneous and is proportional to the applied current
(the `F' phase). This is followed by further depolarization
5. Some internodal conductances can be studied using that develops slowly over some tens of milliseconds as the
`threshold electrotonus' current spreads to and depolarizes the internodal membrane
(the `S1' phase, producing the upward de¯ections in Fig. 2).
The technique that provides most insight into internodal The threshold decrease (i.e. the extent of depolarization)
conductances in human subjects in vivo is `threshold elec- reaches a peak ,20 ms after the onset of the current
trotonus' (Bostock and Baker, 1988). This term was coined pulse, dependent on its strength, and then threshold starts
for the changes in threshold produced by long-lasting DC to return slowly towards the control level. This lessening of
pulses (Fig. 2), because under most circumstances, the the degree of depolarization is due to activation of a hyper-
changes in threshold largely parallel the underlying electro- polarizing conductance with slow kinetics and is called
tonic changes in membrane potential. Conventionally, `S2.' The use of K 1 channel blocking agents indicates
threshold electrotonus is plotted such that an increase in that the slow accommodative process is due to activation
excitability (i.e. a threshold reduction) produces an upwards of slow K 1 channels, which are located on both the node

Fig. 2. Threshold electrotonus of sensory and motor axons in the ulnar nerve at the wrist. Changes in the threshold stimulus current for the target potential
produced by polarizing currents lasting 300 ms, the intensity of the polarizing currents being 40% of threshold in the depolarizing direction and 40% and 80%
of threshold in the hyperpolarizing direction for both sets of ®bres. A reduction in threshold produced by depolarizing current is plotted as an upward de¯ection,
so that the waveforms look like the underlying electronic potentials. The different components of threshold electrotonus are labelled on panel A. The only
difference in threshold electrotonus of sensory and motor ®bres is in S3, the inward recti®cation due to IH. Each panel shows the mean data for 8 subjects ^ 1
standard error. From Bostock et al. (1994), with permission.
D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585 1579

and the internode (Baker et al., 1987; Bostock and Baker, 7. Excitability properties re¯ect membrane potential
1988; Baker and Bostock, 1989). When the DC pulse is
terminated, threshold increases rapidly, and there is then a It is not possible to measure membrane potential in intact
slow overshoot before it gradually recovers to the control human axons, but it is possible to obtain indirect evidence
level. The slow overshoot is due to passive repolarization of about membrane potential by studying excitability indices
the internodal membrane, while the ®nal recovery to the that are dependent on membrane potential. Each of the
control level is due to the deactivation of the K 1 conduc- indices below can be altered by factors other than membrane
tance. potential, but it is a reasonable conclusion that membrane
potential is different when these indices undergo changes in
5.2. There is accommodation to long-lasting the appropriate direction and by the appropriate extent.
hyperpolarizing current pulses

With long-lasting hyperpolarizing DC pulses, there is 7.1. Threshold


again a fast threshold change that increases threshold, During many acute manoeuvres, the stimulus current
proportionally to the applied current, analogous to the necessary to activate an axon or to produce a compound
comparable phase with depolarizing currents (the `F' potential of ®xed size (i.e. the threshold for the potential)
phase). Then threshold continues to increase as the hyper- can be used as a surrogate marker of membrane potential.
polarization spreads to the internode (producing the down-
ward de¯ections in Fig. 2). This `S1' phase is not the mirror
image of the S1 phase with depolarizing currents because
hyperpolarization closes internodal K 1 channels (and this
will increase internodal resistance and the size of S1) while
depolarization activates internodal K 1 channels (and this
will limit the size of S1). At ,100 ms after the start of
the hyperpolarizing current pulse, S1 reaches a maximum
and threshold begins to decrease towards the control level.
This accommodative `S3' phase produces inward recti®ca-
tion and is due to activation of IH. Although IH has slow
activation and deactivation kinetics, it is activated and
affects threshold earlier than 100 ms (where S1 peaks):
without IH, the hyperpolarizing threshold increase would
be even greater. On termination of the hyperpolarizing DC
pulse, threshold rapidly decreases and then slowly under-
shoots the resting threshold until IH deactivates and the slow
K 1 conductance reactivates.

6. Excitability ¯uctuates after an action potential

Following an action potential, large myelinated axons are


absolutely refractory for 0.5±1.0 ms, during which they
cannot generate another action potential no matter how
strong the depolarizing stimulus. The axons are then rela-
tively refractory for some 3±4 ms, during which a stronger
than normal depolarizing stimulus is required to generate an
action potential. As refractoriness subsides, axons pass into
a phase of supernormality (or superexcitability) during
which the stimulus necessary to generate an action potential Fig. 3. The recovery of excitability of sensory and motor axons in the
median nerve at the wrist following a single supramaximal conditioning
is less than at rest, the mechanisms for which are discussed
stimulus. Panel A shows the data for compound sensory action potentials of
later (Fig. 3). This phase peaks at ,7 ms in cutaneous 3 different sizes. Panel B shows the equivalent data for motor axons. In this
afferents of the sural and median nerves and in motor ®gure a reduction in excitability is plotted upwards. At short-conditioning
axons of the peroneal and median nerves (Kiernan et al., test intervals, the axons fall within the relatively refractory period, and
1996; Lin et al., 2000a; Kuwabara et al., 2000). Thereafter threshold is increased. At 3±4 ms, axons enter the supernormal period,
and less current is required to activate them, such that threshold is lower
axons become less excitable again as they pass into the late
than control for ,20 ms, when axons enter the late subnormal period. Note
subnormal (or subexcitable) period, returning to resting that the threshold excursions are smaller for sensory ®bres than for motor,
excitability slowly, 100±150 ms after the conditioning but that the changes are similar for compound potentials of different size.
discharge. From Kiernan et al. (1996), with permission.
1580 D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585

As the membrane depolarizes, the threshold current neces- this alters Na 1 channel inactivation. It is generally assumed
sary to produce the target potential decreases, and as the that, at resting membrane potential, some 30% of Na 1 chan-
membrane hyperpolarizes threshold increases. However, nels are inactivated (Hodgkin and Huxley, 1952; see
because threshold varies considerably between subjects Schwarz et al., 1995). Hyperpolarizing shifts in membrane
due to geometric factors, resting threshold does not provide potential remove this `resting' inactivation and decrease the
useful information about resting membrane potential. Such extent of refractoriness. Conversely, depolarizing shifts in
information can instead be obtained by comparing threshold membrane potential will increase the extent of inactivation
before and after a conditioning nerve impulse, or before and and thereby increase refractoriness (Fig. 4). Unlike most
during the application of a polarizing current. other excitability parameters, refractoriness is extremely
Apart from the issue of intersubject variability, changes temperature sensitive, however measured (Burke et al.,
in excitability do not always parallel changes in membrane 1998; Kiernan et al., 2001b). It is therefore essential to
potential. Membrane depolarization reduces the voltage take temperature into account if refractoriness is to be
change necessary to activate Na 1 channels, but it also inac- used as an index of membrane potential.
tivates Na 1 channels, reducing the number available for
action potential generation. The greater the depolarization, 7.3. Supernormality
the more important the effect on Na 1 channel inactivation
When the node depolarizes to produce an action potential,
becomes, so that with progressive depolarization, such as
current is stored on the internodal membrane and then
occurs during ischaemia, the reduction in threshold does not
discharges through high resistance pathways under or
continue inde®nitely, but reaches a limit and then starts to
through the myelin. This produces the negative (or depolar-
increase, ultimately resulting in depolarizing block. In addi-
izing) afterpotential that is responsible for the phase of
tion, ischaemia produces a voltage-independent block of
supernormal excitability (Barrett and Barrett, 1982; Blight,
Na 1 channels, possibly due to intracellular acidosis, and
1985; Bowe et al., 1987). The extent of supernormality
this accentuates the discrepancy between excitability (or
varies with membrane potential because this alters the resis-
threshold) and membrane potential (Grosskreutz et al.,
tance of the paranodal membrane (by voltage-dependent
1999, 2000). A further example is the increase in axonal
effects on paranodal K 1 channels; David et al., 1992,
excitability that occurs during hyperventilation, which is
1995). However, factors that alter myelin resistance (such
probably due to changes in extracellular pH (rather than
as paranodal demyelination) should also alter the extent of
extracellular Ca 21 concentration) and with which changes
supernormality.
in membrane potential are secondary rather than primary
Depolarization of the internodal membrane opens para-
(Mace®eld and Burke, 1991; Mogyoros et al., 1997b).
nodal K 1 channels and this reduces the resistance of the
internodal membrane, and thereby the size of the depolariz-
7.2. Refractoriness ing afterpotential and the extent of supernormality (Fig. 4).
Hyperpolarization closes paranodal K 1 channels, increases
It is impossible in human subjects to measure the extent
internodal membrane resistance, and thereby increases the
or duration of absolute refractoriness because there is a
depolarizing afterpotential and the extent of supernormality.
limitation on the stimulus that can be delivered to a patient.
Supernormality and refractoriness are both more sensitive
Relative refractoriness has been measured in two ways: as
than the resting threshold to changes in membrane potential,
the increase in current required to produce the target poten-
but whereas the potential dependence of refractoriness
tial at a ®xed conditioning-test interval, usually 2 ms (e.g.
depends primarily on the properties of nodal Na 1 channels,
Burke et al., 1998; Bostock et al., 1998), or as the duration
the potential dependence of supernormality depends primar-
of the relatively refractory period (normally 3±4 ms at skin
ily on paranodal K 1 channels.
temperatures of 32±348C; Kiernan et al., 2000, 2001b). The
former is well-suited for following the evolution of a 7.4. Strength-duration time constant
number of indices of axonal excitability during experimen-
tal manoeuvres, such as ischaemia and membrane polariza- The strength-duration time constant (t SD) is a measure of
tion (Mogyoros et al., 1997b; Burke et al., 1998). However, the rate at which the threshold current for a target potential
extreme hyperpolarization can cause supernormality to declines as stimulus duration is increased, and in the formu-
intrude into this conditioning-test interval (Burke et al., lation of Weiss (1901) it equates to chronaxie. Rheobase is
1998), just as extreme depolarization can cause refractori- the threshold for a stimulus that can be in®nitely long, and
ness to intrude into conditioning-test intervals normally both are properties of the nodal membrane. The voltage-
associated with maximal supernormality (Mogyoros et al., dependence of these properties is due to conductances that
1997b; Burke et al., 1998). These problems do not occur are active near threshold, particularly a rapidly activating
when refractoriness is estimated using the duration of the depolarizing conductance that inactivates slowly, if at all
relatively refractory period, but this requires measurements (Bostock and Rothwell, 1997). These properties are those
at multiple conditioning-test intervals. of persistent Na 1 currents, and computer modelling of the
Refractoriness changes with membrane potential because behaviour of human sensory and motor axons suggests that
D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585 1581

Fig. 4. The effects of changing membrane potential on threshold electrotonus (A) and the recovery cycle (B). With background depolarization, there is a
collapsing in (`fanning in') of the threshold electrotonus waveform (panel A) and an increase in refractoriness and a decrease in supernormality (panel B). With
background hyperpolarization, the reverse changes occur. In each panel, the behaviour at rest is the middle trace, there being 4 levels of background
depolarization and 4 of hyperpolarization. From Kiernan and Bostock (2000), with permission.

these currents constitute ,2.5% of the total Na 1 current on measured the effects of intermittent DC polarizing currents
sensory axons and ,1% on motor axons (Bostock and (Burke et al., 1998) or of continuous DC polarizing current
Rothwell, 1997). Increasing the fraction of Na 1 current and ischaemia on axonal excitability (Kiernan and Bostock,
that is persistent (or depolarizing the node) will produce a 2000), for sensory and motor axons respectively. In the
longer t SD and lower rheobase. The longer t SD of sensory latter study, a depolarizing current of 1 mA (estimated to
axons is accompanied by a lower rheobase (,50%) than in depolarize the axons by about 4 mV) resulted in large
motor axons, and this difference in rheobase results in less changes in the relatively refractory period (increased by
current being required to maintain conduction in critically 37%), supernormality (decreased by 82%), and threshold
impaired sensory axons than equivalently impaired motor electrotonus (fanning in by 18% in the depolarizing phase
axons. and 71% in the hyperpolarizing phase). There was a smaller
change in strength-duration time constant (increased by
7.5. Threshold electrotonus 5%). Similar magnitudes of change in excitability were
shown when membrane potential was altered indirectly
There are substantial changes in threshold electrotonus
using ischaemia. The patterns of change in these different
when axons are depolarized or hyperpolarized (Fig. 4; see
excitability parameters should provide a useful indication of
Baker and Bostock, 1989; Horn et al., 1996; Kaji, 1997;
changes in resting membrane potential in patients.
Kiernan and Bostock, 2000). With background membrane
hyperpolarization, there is an increase in the S1 phase in
both the depolarizing and hyperpolarizing directions, 8. Axons are not identical
because the resistance of the internodal membrane is
increased due to the closure of paranodal and internodal The sensitivity of an axon to stresses such as ischaemia
K 1 channels. This has been termed `fanning out' by Kaji differs along the course of the axon (Bostock et al., 1991a)
(1997). Conversely, background membrane depolarization but few biophysical differences have been identi®ed so far at
decreases the resistance of the internode because it activates different sites along the same nerve (Mogyoros et al., 1999;
paranodal and internodal K 1 channels, thereby decreasing Kuwabara et al., 2000). However, there is good evidence for
S1 with both depolarizing and hyperpolarizing currents, and subtle biophysical differences between axons of different
producing an appearance of `fanning in' (Fig. 4). modality, perhaps because their discharge rates and
discharge patterns differ.
7.6. Excitability parameters and membrane potential
8.1. Differences between sensory and motor axons
The extent of the dependence of each of these parameters
on membrane potential has been explored in studies that When compared with upper-limb cutaneous afferents,
1582 D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585

upper-limb motor axons undergo less refractoriness, super- when trains of impulses are conducted, but only the slow
normality and late subnormality following a single changes due to the late subnormal phase are cumulative.
discharge (Kiernan et al., 1996; see Fig. 3), have a shorter This causes axonal hyperpolarization which is maximal
strength-duration time constant and a higher rheobase with some 10±20 impulses in the train and which decays
(Panizza et al., 1994; Mogyoros et al., 1996; Bostock and back to resting level over some 100±150 ms (Bergmans,
Rothwell, 1997), and probably express a lesser persistent 1970; Taylor et al., 1992; Miller et al., 1995). This decrease
Na 1 conductance (Bostock and Rothwell, 1997) and less in excitability was termed H1 by Bergmans (1970) because
inward recti®cation (IH) (Bostock et al., 1994; see Fig. 2). it seems to correlate with the ®rst positive afterpotential (P1)
As a result of some of these differences, it has been argued described by Gasser (1935). In median cutaneous afferents,
that motor axons are more likely to undergo conduction H1 increases threshold by some 40%, but for sural afferents
block than cutaneous afferents but less prone to become the threshold increase is less, some 20±30% (Lin et al.,
ectopically active (Burke et al., 1997; Bostock et al., 2000a). Given that H1 is largely due to a slow K 1 conduc-
1998). For example, the difference in IH would render tance (Baker et al., 1987), this provides some evidence for a
motor axons less protected from hyperpolarization and, difference in nodal slow K 1 channels for median and sural
indeed, there is evidence that activity-dependent hyperpo- afferents. The hyperpolarization caused by H1 is probably
larization is greater in motor axons than sensory for equiva- too brief to cause conduction block, but it is suf®ciently
lent discharge rates (Vagg et al., 1998). When action strong that it could act as a rate-limiting mechanism for
potential generation is precarious, the higher rheobase of diseased axons.
motor axons suggests that motor axons may require more
current to keep them conducting than sensory axons. 9.2. Axons undergo long-lasting hyperpolarization
following prolonged impulse trains
8.2. Differences between different sensory axons and
different motor axons Impulse trains that last a few seconds will produce an
accumulation of Na 1 ions inside the axon and K 1 ions
Comparison of measurements of late subnormality and outside the axon, and this altered distribution stimulates
threshold electrotonus suggest that cutaneous afferents in the Na 1/K 1 pump (Fig. 5). However, the pump is `electro-
the sural nerve have lesser nodal and internodal slow K 1 genic': the ion swap is not balanced, and pump activity
conductances and less IH than cutaneous afferents in the therefore results in axonal hyperpolarization. The term
median nerve (Lin et al., 2000a,b). These differences `H2' was coined by Bergmans (1970) to describe the result-
could predispose sural afferents to undergo dysfunction ing threshold change because it seems analogous to the P2
before median afferents, and this may be a factor in why positive afterpotential described by Gasser (1935).
acquired polyneuropathies are usually more severe distally In human axons, the membrane hyperpolarization
in the lower limbs. Peroneal motor axons probably also increases in depth with the number of pulses in the train
express a lesser slow K 1 conductance than median motor and then reaches a plateau, further impulses prolonging
axons but IH appears to be similar (Kuwabara et al., 2000, the time that threshold remains elevated following cessation
2001). of the impulse train (Bergmans, 1970; Bostock and Berg-

9. Conducting trains of impulses alters axonal


excitability

As illustrated in Fig. 3, the generation of an action poten-


tial at the node of Ranvier sets up a sequence of oscillating
changes in membrane potential which, following a single
action potential, are commonly referred to as the recovery
cycle. Trains of impulses result in summation of these
effects, compensatory mechanisms to correct the altered
distribution Na 1 and K 1 ions on either side of the
membrane (e.g. activation of the Na 1/K 1 pump) and the
secondary effects of processes designed to minimize unto-
ward effects of pump activation (e.g. activation of IH to
reduce the pump-induced hyperpolarization).

9.1. Axons hyperpolarize when they conduct brief trains of


impulses

The changes induced during the recovery cycle also occur Fig. 5. Mechanisms of activity-dependent conduction block.
D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585 1583

mans, 1994; Kiernan et al., 1997a). Other processes become ,40% (Fig. 6; Vagg et al., 1998). Importantly the hyperpo-
more important the longer the impulse train, particularly larization may take some 10±15 min to decay to pre-
with unnaturally long high-frequency trains lasting 5±30 contraction levels on cessation of the voluntary effort. The
min (Bergmans, 1970; Applegate and Burke, 1989; Bostock extent and duration of the activity-dependent hyperpolariza-
et al., 1991b; Bostock and Bergmans, 1994; Kiernan et al., tion is insuf®cient to jeopardize normal impulse traf®c in
1997a,b). These changes can result in ectopic impulse activ- healthy axons, but it can trigger conduction block in demye-
ity in sensory and motor axons but are outside the scope of linated axons of patients with chronic in¯ammatory demye-
this review. linating polyneuropathy and multifocal motor neuropathy
Natural activity can produce substantial axonal hyperpo- (Fig. 5; see Cappelen-Smith et al., 2000; Kaji et al., 2000).
larization (Fig. 6), and this appears to be greater for motor
axons than sensory, probably because the former have less
IH (see earlier). A maximal voluntary contraction of abduc- 10. Of what clinical value are measurements of axonal
tor pollicis brevis lasting only 15 s increases the threshold of excitability?
median motor axons (measured using 1-ms test pulses) by
15±20% and contraction for 1 min increases threshold by As a clinical tool, measurements of axonal excitability are
still in their infancy, the physiological and technical factors
that can affect the measurements are being explored, clini-
cally useful protocols are being developed, and their varia-
bility in normal subjects is being assessed. To be useful
clinically, testing should assess multiple indices relatively
quickly, so that the procedures can be used, when appropri-
ate, to supplement conventional nerve conduction studies.
This rationale underlies the development of a rapid testing
protocol that assesses stimulus/response curves, strength-
duration properties, threshold electrotonus and recovery
cycle for motor axons in ,10 min (Kiernan et al., 2000).
A similar but, of necessity, slightly longer protocol has been
developed for sensory axons (Kiernan et al., 2001d).
However, at the moment, studies of axonal excitability
have been used in only a limited number of disease
processes (Table 2), generally to provide insight into the
underlying pathophysiology, rather than diagnosis. For
example, in patients with diabetic neuropathy, the decrease
in threshold that occurs during ischaemia and the increase in
threshold that occurs on release of ischaemia are less than in
healthy nerve (Weigl et al., 1989; Strupp et al., 1990). This
is the basis of the so-called `ischaemic resistance' of
diabetic nerve and, as a result, diabetic patients experience
fewer paraesthesiae during and after ischaemic insults. The
threshold abnormalities correlate with the degree of diabetic
control. In addition, studies using threshold electrotonus
have demonstrated that there is less IH in diabetic sensory
and motor axons than normal (Horn et al., 1996). In patients
with motor neurone disease, there is evidence for decreased
K 1 conductances (Bostock et al., 1995; Horn et al., 1996)
and a longer t SD, perhaps indicating increased INap on motor
axons (Mogyoros et al., 1998a,b). Both of these changes
would increase axonal excitability. A recent ®nding with
Fig. 6. Activity-dependent hyperpolarization of motor axons following
contractions of different duration. The ®gure shows the changes in thresh- major implications for diagnostic practice has been the
old, of median motor axons at the wrist following maximal voluntary demonstration that conventional measurements of conduc-
contractions of abductor pollicis brevis for 15 s, 30 s and 60 s. The contrac- tion block in demyelinating neuropathies, using low stimu-
tions began at 5 min on the horizontal axis, and resulted in an increase in the lation rates, are likely to underestimate the true extent of
current required to produce the target compound muscle action potential of
conduction block experienced by the patient (Cappelen-
the thenar muscles. Following the 60-s contraction, the increase in current
was ,40% and it took 10±15 min to decay to control values. A smaller but Smith et al., 2000; Kaji et al., 2000). Such measurements
signi®cant increase followed the 15-s contraction. Mean data for 6 subjects. would be improved by testing before and after a standar-
From Vagg et al. (1998), with permission. dized voluntary contraction.
1584 D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585

Acknowledgements voltage dependence of axonal excitability in human cutaneous afferents.


Brain 1998;121:1975±1983.
Caldwell JH, Shaller KL, Lasher RS, Peles E, Levinson SR. Sodium chan-
This work was supported by the National Health and nel Na(v)1.6 is localized at nodes of Ranvier, dendrites, and synapses.
Medical Research Council of Australia. MCK was recipient Proc Natl Acad Sci USA 2000;97:5616±5620.
of a C.J. Martin/R.G. Menzies Travelling Fellowship of the Cappelen-Smith C, Kuwabara S, Lin CS-Y, Mogyoros I, Burke D. Activity-
National Health and Medical Research Council of Australia. dependent hyperpolarization and conduction block in chronic in¯am-
matory demyelinating polyneuropathy. Ann Neurol 2000;48:826±832.
Culp WJ, Ochoa J, editors. Abnormal nerves and muscles as impulse
generators Oxford: Oxford University Press, 1980.
References David G, Barrett JN, Barrett EF. Evidence that action potentials activate an
internodal potassium conductance in lizard myelinated axons. J Physiol
(Lond) 1992;445:277±301.
Applegate C, Burke D. Changes in excitability of human cutaneous affer-
David G, Modney B, Scappaticci KA, Barrett JN, Barrett EF. Electrical and
ents following prolonged high-frequency stimulation. Brain
morphological factors in¯uencing the depolarizing after-potential in rat
1989;112:147±164.
and lizard myelinated axons. J Physiol (Lond) 1995;489:141±157.
Baker MD. Axonal ¯ip-¯ops and oscillators. Trends Neurosci
Gasser HS. Changes in nerve potentials produced by rapidly repeated
2000;23:514±519.
stimuli and their relation to the responsiveness of nerve to stimulation.
Baker M, Bostock H. Depolarization changes the mechanism of accommo-
Am J Physiol 1935;111:35±50.
dation in rat and human motor axons. J Physiol (Lond) 1989;411:545± Goldin AL. Diversity of mammalian voltage-gated sodium channels. Ann
561. N Y Acad Sci 1999;868:38±50.
Baker M, Bostock H. Low-threshold, persistent sodium current in rat large Goldin AL, Barchi RL, Caldwell JH, Hofmann F, Howe JR, Hunter JC,
dorsal root ganglion neurons in culture. J Neurophysiol 1997;77:1503± Kallen RG, Mandel G, Meisler MH, Netter YB, Noda M, Tamkun MM,
1513. Waxman SG, Wood JN, Catterall WA. Nomenclature of voltage-gated
Baker M, Bostock H. Inactivation of macroscopic late Na 1 current and sodium channels. Neuron 2000;28:365±368.
characteristics of unitary late Na1 currents in sensory neurons. J Grosskreutz J, Lin CS-Y, Mogyoros I, Burke D. Changes in excitability
Neurophysiol 1998;80:2538±2549. indices of cutaneous afferents produced by ischaemia in human
Baker M, Bostock H, Grafe P, Martius P. Function and distribution of 3 subjects. J Physiol (Lond) 1999;518:301±314.
types of rectifying channel in rat spinal root myelinated axons. J Physiol Grosskreutz J, Lin CS-Y, Mogyoros I, Burke D. Ischaemic changes in
(Lond) 1987;383:45±67. refractoriness of human cutaneous afferents under threshold-clamp
Barrett EF, Barrett JN. Intracellular recording from vertebrate myelinated conditions. J Physiol (Lond) 2000;523:807±815.
axons: mechanism of depolarizing afterpotential. J Physiol (Lond) Hanauske AR, Schilling T, Heinrich B, Kau R, Herzog M, Quasthoff S,
1982;323:117±144. Bochtler H, Diergarten K, Rastetter J. Clinical phase I study of Pacli-
Bergmans J. The physiology of single human nerve ®bres, Louvain: taxel followed by Cisplatin in advanced head and neck squamous cell
Vander, 1970. carcinoma. Semin Oncol 1995;22:35±39.
Blight AR. Computer simulation of action potentials and afterpotentials in Hodgkin AL, Huxley AF. A quantitative description of membrane current
mammalian myelinated axons: the case for a lower resistance myelin as its application to conduction and excitation in nerve. J Physiol (Lond)
sheath. Neuroscience 1985;15:13±31. 1952;117:500±544.
Bostock H, Baker M. Evidence for two types of potassium channel in Horn S, Quasthoff S, Grafe P, Bostock H, Renner R, Schrank B. Abnormal
human motor axons in vivo. Brain Res 1988;462:354±358. axonal inward recti®cation in diabetic neuropathy. Muscle Nerve
Bostock H, Bergmans J. Post-tetanic excitability changes and ectopic 1996;19:1268±1275.
discharges in a human motor axon. Brain 1994;117:913±928. Inglis JT, Leeper JB, Wilson LR, Gandevia SC, Burke D. The development
Bostock H, Grafe P. Activity-dependent excitability changes in normal and of conduction block in single human axons following a focal nerve
demyelinated rat spinal root axons. J Physiol (Lond) 1985;365:239± injury. J Physiol (Lond) 1998;513:127±133.
257. Kaji R. Physiological and technical basis of peripheral nerve and moto-
Bostock H, Rothwell JC. Latent addition in motor and sensory ®bres of neuron testing. In: Kimura J, Kaji R, editors. Physiology of ALS and
human peripheral nerve. J Physiol (Lond) 1997;498:277±294. related diseases, Amsterdam: Elsevier Science, 1997. pp. 15±41.
Bostock H, Baker M, Grafe P, Reid G. Changes in excitability and accom- Kaji R, Bostock H, Kohara N, Murase N, Kimura J, Shibasaki H. Activity-
modation of human motor axons following brief periods of ischaemia. J dependent conduction block in multifocal motor neuropathy. Brain
Physiol (Lond) 1991a;441:513±535. 2000;123:1602±1611.
Bostock H, Baker M, Reid G. Changes in excitability of human motor Kiernan MC, Bostock H. Effects of membrane polarization and ischaemia
axons underlying post-ischaemic fasciculations: evidence for two stable on the excitability properties of human motor axons. Brain
states. J Physiol (Lond) 1991b;441:537±557. 2000;123:2542±2551.
Bostock H, Burke D, Hales JP. Differences in behaviour of sensory and Kiernan MC, Mogyoros I, Burke D. Differences in the recovery of excit-
motor axons following release of ischaemia. Brain 1994;117:225±234. ability in sensory and motor axons of human median nerve. Brain
Bostock H, Sharief MK, Reid G, Murray NMF. Axonal ion channel 1996;119:1099±1105.
dysfunction in amyotrophic lateral sclerosis. Brain 1995;118:217±225. Kiernan MC, Hales JP, Gracies J-M, Mogyoros I, Burke D. Paraesthesiae
Bostock H, Cikurel K, Burke D. Threshold tracking techniques in the study induced by prolonged high frequency stimulation of human cutaneous
of human peripheral nerve. Muscle Nerve 1998;21:137±158. afferents. J Physiol (Lond) 1997a;501:461±471.
Bowe CM, Kocsis JD, Waxman SG. The association of the supernormal Kiernan MC, Mogyoros I, Hales JP, Gracies J-M, Burke D. Excitability
period and the depolarizing afterpotential in myelinated frog and rat changes in human cutaneous afferents induced by prolonged repetitive
sciatic nerve. Neuroscience 1987;21:585±593. axonal activity. J Physiol (Lond) 1997b;500:255±264.
Burke D, Kiernan MC, Mogyoros I, Bostock H. Susceptibility to conduc- Kiernan MC, Mogyoros I, Burke D. Conduction block in carpal tunnel
tion block: differences in the biophysical properties of cutaneous affer- syndrome. Brain 1999;122:933±941.
ents and motor axons. In: Kimura J, Kaji R, editors. Physiology of ALS Kiernan MC, Burke D, Andersen KV, Bostock H. Multiple measures of
and related diseases, Amsterdam: Elsevier Science, 1997. pp. 43±53. axonal excitability: a new approach in clinical testing. Muscle Nerve
Burke D, Mogyoros I, Vagg R, Kiernan MC. Quantitative description of the 2000;23:399±409.
D. Burke et al. / Clinical Neurophysiology 112 (2001) 1575±1585 1585

Kiernan MC, Baker MD, Bostock H. Tetrodotoxin-sensitive and -resistant properties and their voltage dependence at different sites along the
late Na 1 currents in adult rat small sensory neurones in culture. J median nerve. Clin Neurophysiol 1999;110:1618±1624.
Physiol (Lond) 2001a;531:82P. Mogyoros I, Bostock H, Burke D. Mechanisms of paresthesias arising from
Kiernan MC, Cikurel K, Bostock H. Effects of temperature on the excit- healthy axons. Muscle Nerve 2000;23:310±320.
ability properties of human motor axons. Brain 2001b;124:816±825. Nordin M, NystroÈm B, Wallin U, Hagbarth KE. Ectopic sensory discharges
Kiernan MC, Hart IK, Bostock H. Excitability properties of motor axons in and paresthesiae in patients with disorders of peripheral nerves, dorsal
patients with spontaneous motor unit activity. J Neurol Neurosurg roots and dorsal columns. Pain 1984;20:231±245.
Psychiatry 2001c;70:56±64. NystroÈm B, Hagbarth KE. Microelectrode recordings from transected
Kiernan MC, Lin CS, Andersen KV, Bostock H. Clinical evaluation of nerves in amputees with phantom limb pain. Neurosci Lett
excitability measures in sensory nerve. Muscle Nerve 2001d in press. 1981;27:211±216.
Kuwabara S, Cappelen-Smith C, Lin CS-Y, Mogyoros I, Bostock H, Burke Ochoa J, TorebjoÈrk HE. Paresthesiae from ectopic impulse generation in
D. Excitability properties of median and peroneal motor axons. Muscle human sensory nerves. Brain 1980;103:835±854.
Nerve 2000;23:1365±1373. Pape HC. Queer current and pacemaker: the hyperpolarization-activated
Kuwabara S, Cappelen-Smith C, Lin CS-Y, Mogyoros I, Burke D. Differ- cation current in neurons. Annu Rev Physiol 1996;58:299±327.
ences in accommodative properties of median and peroneal motor Panizza M, Nilsson J, Roth BJ, Rothwell J, Hallett M. The time constants of
axons. J Neurol Neurosurg Psychiatry 2001;70:372±376. motor and sensory peripheral nerve ®bers measured with the method of
Lance JW, Burke D, Pollard J. Hyperexcitability of motor and sensory latent addition. Electroenceph clin Neurophysiol 1994;93:147±154.
neurons in neuromyotonia. Ann Neurol 1979;5:523±532. Reid G, Scholz A, Bostock H, Vogel W. Human axons contain at least 5
Lin CS-Y, Mogyoros I, Burke D. Recovery of excitability of cutaneous types of voltage-dependent potassium channels. J Physiol (Lond)
afferents in the median and sural nerves following activity. Muscle 1999;518:681±696.
Nerve 2000a;23:763±770. Ritchie JM. Physiology of axons. In: Waxman SG, Stys PK, Kocsis JD,
Lin CS-Y, Mogyoros I, Kuwabara S, Cappelen-Smith C, Burke D. Accom- editors. The axon, Oxford: Oxford University Press, 1995. pp. 68±96.
modation to depolarizing and hyperpolarizing currents in cutaneous Schilling T, Heinrich B, Kau R, Herzog M, Quasthoff S, Diergarten K,
afferents of the human median and sural nerves. J Physiol (Lond) Rastetter J, Hanauske AR. Paclitaxel administered over 3h followed
2000b;529:483±492. by cisplatin in patients with advanced head and neck squamous cell
Mace®eld G, Burke D. Paraesthesiae and tetany induced by voluntary carcinoma: a clinical phase I study. Oncology 1997;54:89±95.
hyperventilation: increased excitability of human cutaneous and Schwarz JR, Reid G, Bostock H. Action potentials and membrane currents
motor axons. Brain 1991;114:527±540. in the human node of Ranvier. P¯uÈgers Arch 1995;430:382±392.
Maddison P, Newsom-Davis J, Mills KR. Strength-duration properties of Strupp M, Bostock H, Weigl P, Piwernetz K, Renner R, Grafe P. Is resis-
peripheral nerve in acquired neuromyotonia. Muscle Nerve tance to ischaemia of motor axons in diabetic subjects due to membrane
1999;22:823±830. depolarization? J Neurol Sci 1990;99:271±280.
Miller TA, Kiernan MC, Mogyoros I, Burke D. Activity-dependent changes Taylor JL, Burke D, Heywood J. Physiological evidence for a slow K 1
in impulse conduction in focal nerve lesion. Brain 1995;118:1217± conductance in human cutaneous afferents. J Physiol (Lond)
1224. 1992;453:575±589.
Mogyoros I, Kiernan MC, Burke D. Strength-duration properties of human Vagg R, Mogyoros I, Kiernan MC, Burke D. Activity-dependent hyperpo-
peripheral nerve. Brain 1996;119:439±447. larization of motor axons produced by natural activity. J Physiol (Lond)
Mogyoros I, Kiernan MC, Burke D. Strength-duration properties of cuta- 1998:919±925.
neous and motor axons in carpal tunnel syndrome. Muscle Nerve Vogel W, Schwarz JR. Voltage-clamp studies on axons: macroscopic and
1997a;20:508±510. single-channel currents. In: Waxman SG, Stys PK, Kocsis JD, editors.
Mogyoros I, Kiernan MC, Burke D, Bostock H. Excitability changes in The axon, Oxford: Oxford University Press, 1995. pp. 257±280.
human sensory and motor axons during hyperventilation and ischaemia. Waxman SG, Cummins TR, Dib-Hajj S, Fjell J, Black JA. Sodium chan-
Brain 1997b;120:317±325. nels, excitability of primary sensory neurons, and the molecular basis of
Mogyoros I, Kiernan MC, Burke D, Bostock H. Ischaemic resistance of pain. Muscle Nerve 1999;22:1177±1187.
cutaneous afferents and motor axons in patients with amyotrophic Weigl P, Bostock H, Franz P, Martius P, MuÈller W, Grafe P. Threshold
lateral sclerosis. Muscle Nerve 1998a;21:1692±1700. tracking provides a rapid indication of ischaemic resistance in motor
Mogyoros I, Kiernan MC, Burke D, Bostock H. Strength-duration proper- axons of diabetic subjects. Electroenceph clin Neurophysiol
ties of sensory and motor axons in amyotrophic lateral sclerosis. Brain 1989;73:369±371.
1998b;121:851±859. Weiss G. Sur la possibilite de rendre comparables entre eux les appareils
Mogyoros I, Lin CS-Y, Dowla S, Grosskreutz J, Burke D. Strength-duration servant aÁ l'excitation eÂlectrique. Arch Ital Biol 1901;35:413±446.

You might also like