Вы находитесь на странице: 1из 10

Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78

Contents lists available at ScienceDirect

Autonomic Neuroscience: Basic and Clinical


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a u t n e u

Review

Visceral afferents — Determinants and modulation of excitability


Michael J. Beyak ⁎
Gastrointestinal Diseases Research Unit (GIDRU), Queen's University, Kingston, Ontario, Canada

a r t i c l e i n f o a b s t r a c t

Article history: An essential property of visceral sensory afferents is to be able to alter their firing properties in response to
Received 25 June 2009 changes in the microenvironment at the level of the sensory ending. Significant progress has been made in
Received in revised form 13 July 2009 recent years in understanding the ionic mechanisms of the regulation of afferent neuronal excitability, and in
Accepted 20 July 2009
identifying the mechanisms by which this can be altered. This article will review some of the recent
developments in the state of knowledge regarding mechanisms of increased excitability after inflammation,
Keywords:
and pharmacological modulation of excitability, concentrating on afferent nerves innervating the GI tract and
Visceral afferent
Ion channels
urinary bladder.
Pharmacology © 2009 Elsevier B.V. All rights reserved.
Excitability

Contents

1. Techniques — recording from afferent nerves . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70


1.1. Afferent nerve fibre recording . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
1.2. Isolated cell recording — intracellular/patch clamp recording. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
2. Ion channel determinants of excitability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
2.1. Voltage gated sodium (NaV) channels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
2.2. Voltage gated potassium (KV) channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
3. Pharmacological modulation of excitability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
4. Ligand gated ion channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
4.1. TRPV channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
4.2. P2X receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72
4.3. Serotonin — 5HT3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72
4.4. GPCRs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72
5. Protease activated receptors — PARs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72
6. Metabotropic glutamate receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
7. Somatostatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
8. GABAB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
9. Satiety hormones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
10. Gasotransmitters NO/H2S. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
11. Chronic alteration in visceral excitability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
11.1. Inflammation and afferent excitability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
11.2. Mechanisms of inflammation induced hyperexcitability — alterations in receptor and ion channel function . . . . . . . . . . . . . . . . 75
12. Visceral hyposensitivity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
13. Summary and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76

At the heart of the function of the sensory neuron is its ability


to change its firing pattern in response to alterations in the micro-
⁎ Departments of Medicine and Physiology, Queen's University, GIDRU Wing, Kingston
General Hospital, 76 Stuart St, Kingston, ON, Canada K7L 2V7. Fax: +1 613 548 2426. environment of the nerve ending. This enables detection of physiolo-
E-mail address: beyakm@queensu.ca. gical stimuli, as well as noxious ones. In addition, afferent neurons,

1566-0702/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.autneu.2009.07.019
70 M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78

including visceral afferents exhibit a great degree of plasticity in their which they will ultimately be used, such as the specialized afferent
ability to respond to a given stimulus. That is to say the response to the ending. Nonetheless, these techniques have given tremendous insight
same stimulus is context dependent, and is influenced by what is into the regulation of sensory neuronal excitability and when com-
happening in the microenvironment at the level of the sensory ending. pared with results obtained from afferent nerve fibre recording ex-
For the purposes of this review “excitability” will be considered to refer periments, have fared reasonably well.
to the degree of response of a neuron in terms of action potential
output to a given stimulus. An increase in excitability is an action 2. Ion channel determinants of excitability
potential in response to a lesser stimulus, or a greater number of action
potentials fired in response to a similar stimulus. This review will 2.1. Voltage gated sodium (NaV) channels
concentrate on selected recent advances in the study of the regulation
of visceral afferent excitability, focusing on afferents innervating the Voltage gated sodium channels are essential in mediating the rapid
gastrointestinal tract and urinary bladder. upstroke of the action potential . The regenerative depolarization,
then further channel activation, and subsequent further depolariza-
1. Techniques — recording from afferent nerves tion result in the rapid upstroke of the action potential (Hille, 2001).
The threshold for activation of the sodium channel is the key in setting
There are a number of experimental approaches that can be taken the action potential voltage threshold. Equally important is the time
to assess visceral afferent excitability, and in broad categories they can dependent inactivation of these channels, terminating the upstroke,
be divided into recordings from nerve fibres (teased single axons or and permitting repolarization of the membrane. Sensory neurons are
multiunit recordings) or recordings from single neuronal cell bodies. relatively unique in that the voltage gated channels present are
limited to few subunits (Waxman et al., 1999b) In visceral sensory
1.1. Afferent nerve fibre recording neurons, many of these are unmyelinated a delta or C fibres, and thus
also carry a unique repertoire of sodium channel alpha subunits.
This technique involves extracellular recording of afferent nerve Voltage gated sodium channels in visceral sensory neurons can be
activity using a wire or suction electrode, and then this signal is broadly categorized into 2 categories based on their sensitivities to the
differentially amplified, giving a readout of individual action potentials puffer fish toxin tetrodotoxin (TTX) (Waxman et al., 1999a). Most
or “spikes”. These types of recordings can be performed in vitro or visceral sensory neurons have both TTX sensitive and TTX resistant
in vivo. The advantage of this approach is that the response of the sodium currents (Beyak et al., 2004; Bielefeldt et al., 2002a,b; Stewart
nerve ending to quasi physiological stimuli can be easily assessed (e.g. et al., 2003; Yoshimura & de Groat, 1997). The alpha subunits
stretch of a hollow viscus, application of mediators to the nerve end- underlying these currents are primarily NaV 1.7 (TTX sensitive) and
ing). In addition, the events recorded are reflective of the behaviour of NaV 1.8 and 1.9 (TTX resistant). NaV 1.7 mediates a TTX sensitive
the specialized sensory ending lying in the wall of the viscus of interest. current with relatively low threshold for activation, rapid inactivation,
The disadvantage of this technique is that the only variable that can be slow repriming kinetics and a hyperpolarized steady state inactivation
assessed is firing frequency. Analysis of the waveform obtained has curve. NaV 1.8 is responsible for a current with a higher threshold for
little usefulness in gaining insight into the biophysical mechanisms of activation, slower inactivation kinetics, rapid repriming kinetics and a
changes in excitability. more depolarized steady state inactivation curve (see Fig. 1A). NaV1.8
channels have been the focus of intense study in visceral sensation
1.2. Isolated cell recording — intracellular/patch clamp recording because of their relatively selective expression in sensory neurons, and
their modulation by inflammatory states. NaV 1.9 results in a relatively
For the reasons indicated above, there needs to be alternate ways of persistent current with a very hyperpolarized availability curve, and is
examining electrical events in more detail at the level of the afferent likely involved in setting the membrane potential and threshold for
neuronal membrane. For this reason a number of investigators have action potential. Relatively few colon projecting afferents exhibit
developed techniques for recording from isolated neuronal cell bodies currents similar to those mediated by NaV 1.9, and this may reflect the
(typically in vitro). Most commonly whole cell patch clamp techniques fact that most colon afferents are peptidergic, rather than IB4 positive
have been used, however some have used sharp microelectrode (Beyak et al., 2004; Roberts et al., 1993) — and the latter have been
intracellular recordings. As sensory neuronal cell bodies lie in ganglia, shown to express NaV 1.9 (Fang et al., 2006) preferentially. Until
these ganglia are often isolated and cells dispersed and maintained in recently the presence of these voltage gated sodium channel subunits
short term culture for electrophysiological recording. Thus isolated in visceral afferents was assumed, based on the expression profiles in
neuronal cells (e.g. Nodose or dorsal root ganglion neurons) can be nonselected DRG and vagal neurons, however a recent paper by King
individually recorded from, and recordings can be made in current et al. has demonstrated the presence of transcript for NaV 1.7, 1.8 and
clamp mode (to measure membrane potential responses) or voltage 1.9 in colon projecting DRG neurons, using laser capture microdissec-
clamp mode (to measure flux of ions across the cell membrane). The tion techniques (King et al., 2009). In addition, in bladder projecting
advantage of these techniques is that very detailed measurements of DRG neurons, the presence of NaV 1.8 and to a lesser degree 1.9 have
individual ionic currents underlying excitability can be made and the been demonstrated, using in situ hybridization techniques (Black et al.,
response of an individual neuron to depolarizing stimuli can be as- 2003).
sessed. The disadvantages are twofold. First, when recording from
randomly selected sensory ganglion neurons, one cannot be comple- 2.2. Voltage gated potassium (KV) channels
tely sure what organs they project to. Many investigators have tackled
this problem through the use of retrogradely transported fluorescent Voltage gated potassium channels are also critical in setting the
dyes, which are applied to the organ of interest prior to cell harvest, excitability of the visceral afferent neuron. They serve to repolarize the
allowing the identification of a specific population of neurons cell membrane, and limit repetitive firing. They can be broadly clas-
projecting to an organ of interest (see Moore et al., 2002; Yoshimura sified based on their inactivation kinetics into inactivating (A-type)
et al., 1996 as examples). The second disadvantage is harder to address. currents or noninactivating IK (or delayed rectifier) currents. IA currents
That is the assumption of these types of recordings is that the events tend to have a more depolarized availability curve, and lower thresholds
occurring at the membrane of the cell body are similar to that at the for inactivation while IK currents have a more depolarized availability
level of the nerve terminal in situ. This may not be entirely correct, as curve and increased thresholds for activation (see Fig. 1B). Reduction
there is likely directed transport of receptors and channels to areas in in A currents results in repetitive firing in visceral sensory neurons,
M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78 71

Fig. 1. Voltage gated channels involved in determining excitability. A) Different voltage gated sodium currents in colon projecting DRG neurons. Left — fast inactivating TTX Sensitive
current. Middle — more slowly inactivating TTX resistant current. Right — ultra slowly inactivating TTX resistant current. Inset — prolonged time course showing little inactivation of
this current over 200 s (modified from Beyak et al., 2004 with permission). B) Typical voltage gated potassium currents in ileum projecting DRG neurons. Top left — total potassium
currents elicited by stepwise depolarizing voltage commands from a holding potential of − 100 mV. Top middle — holding potential of − 60 mV isolates the sustained IK current. Top
right — digital subtraction of the two previous currents reveals IA an inactivating potassium current. Bottom left — total potassium currents from holding potential of − 100 mV.
Middle — currents in the presence of 1 mM 4-aminopyridine, a blocker of IA currents. Bottom right — digital subtraction reveals the presence of IA. Note similarity of these currents to
those obtained in the above 3 panels (modified from Stewart et al., 2003 with permission).

highlighting the importance of this channel in regulating visceral sen- reviews for a more comprehensive list (Beyak et al., 2006a; Blackshaw
sory neuronal excitability (Moore et al., 2002). Decreases in IK current & Gebhart, 2002; Blackshaw et al., 2007).
would be expected to broaden the action potential, and possibly to
depolarize the membrane, as there is likely some of this current active at 4. Ligand gated ion channels
around resting potentials. The molecular determinants of these currents
are likely complex, as studies in unselected DRG neurons indicate Ligand gated channels, as the name suggests, consist of a membrane
expression of numerous KV subunits, many of which underlie these receptor — channel complex, that when activated by ligand binding,
types of currents (Kim et al., 2002; Park et al., 2003; Yang et al., 2004). results in the flux of ions across the cell membrane, thus resulting in
In addition to IA and IK, calcium activated potassium currents also alterations in membrane potential. This can depolarize the membrane
likely play a role in visceral afferent excitability. These currents are resulting in a so called generator potential, and thus activate voltage
likely involved in the afterhyperpolarization and in responses to a gated sodium channels and an action potential.
variety of mediators. The presence of these currents has not been
demonstrated in DRG or neurons projecting to the viscera, however 4.1. TRPV channels
nodose neurons appear to have functional calcium activated potas-
sium channels (Hay & Kunze, 1994a,b). TRPV channels (transient receptor potential vanilloid) are a family
of ligand gated channels and are a member of the TRP family of
3. Pharmacological modulation of excitability channels including the TRPC (canonical TRPs), TRPA (ankyrin), TRPM
(melastatin) and TRPPs (polycystin) members (for recent review see
Visceral afferents display responsiveness to a wide range of endog- Patapoutian et al., 2009). The TRPVs, in particular TRPV1 and TRPV4
enous and exogenous ligands. This property has been termed by have been the focus of a number of interesting studies of visceral
Kirkup et al. as “promiscuous chemosensitivity. (Kirkup et al., 2001)” afferents in recent years. Activation of these channels results in an
In simple categories, the receptors mediating these actions can be inward nonselective cation current. With regard to TRPV1, this is the
classed into ligand gated ion channels and membrane receptors that receptor for capsaicin, the pungent ingredient in chili pepper. How-
activate downstream signaling cascades (e.g. GPCRs), that ultimately ever it is activated by a number of other physical and chemical stimuli
impact on the ion channel determinants of excitability discussed such as acid, heat and likely mechanical forces. Deletion of TRPV1
above (e.g. via channel phosphorylation). As the list of mediators results in diminished afferent responses to mechanical stimuli and
potentially impacting on the function of visceral afferents is immense, acid in the intestine (Rong et al., 2004), stomach (Bielefeldt & Davis,
I will limit the discussion to those examples from the recent liter- 2008), colon (Sugiura et al., 2007; Jones et al., 2005) and urinary
ature, and those receptors that may be important in chronic alter- bladder (Daly et al., 2007) (see Fig. 2). In isolated colon projecting
ations in afferent sensitivity. The reader is otherwise directed to other DRG neurons TRPV mediated acid induced currents as well (Sugiuar
72 M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78

Fig. 2. TRPV1 is involved in afferent mechanosensitivity in the jejunum, colon and urinary bladder. A) Example of jejunal afferent nerve responses to ramp distention in wildtype and
TRPV1−/− mice. Mechanosensitivity is attenuated in the TRPV1−/− animals (modified from Rong et al., 2004 with permission). B) Example of responses of bladder afferents to
ramp distention in wildtype and TRPV1 −/− mice. Similar to the jejunum, there is a significant attenuation of responses to distention in the knockout animals (modified from Daly et
al., 2007 with permission). C) In the colon, deletion of TRPV1 or ASIC3 genes results in attenuation of responses of muscular mucosal afferents to circular stretch (Modified from Jones
et al., 2005 with permission).

et al., 2004). TRPV4 is also a mechanosensitive channel, that is sen- characteristic of the 5-HT3 receptor (Keating et al., 2008). The
sitive to heat (noxious). It also plays a critical role in mechanosensa- importance of this receptor in visceral afferent function is highlighted
tion in the colon and upper GI tract (Brierley et al., 2008). In addition by the use of 5-HT3 antagonists in the treatment of both visceral pain
these channels can be modulated by activation of other receptors such states (e.g. alosetron for IBS) and emesis (ondansetron and granisetron
as PARs (Sipe et al., 2008) (see below) and their function altered by for chemotherapy and postoperative emesis). Furthermore these
chronic inflammation. actions of 5-HT, particularly those on vagal afferents likely underlie
some of the nutrient sensing functions of these afferent nerves (Li
4.2. P2X receptors et al., 2004; Zhu et al., 2001).

ATP, in addition to being the intracellular energy molecule, also 4.4. GPCRs
is known to function as an extracellular signaling molecule. It acts on
two classes of receptors, P2Y (which are GPCRs) and P2X (ligand gated There are literally dozens of G-protein coupled receptor systems that
channels. P2X receptors have been shown to be present in colonic (Xu can increase or decrease afferent excitability. As a detailed review of all
et al., 2008), gastric (Castelucci et al., 2003; Dang et al., 2005; McIlwrath of these is beyond the scope of this article, and is handled in a variety of
et al., 2009) intestinal (Kirkup et al., 1999) and bladder afferent neurons previous reviews, we will focus here on advances made in recent years
(Dang et al., 2008). ATP is a potent stimulus to afferent firing in all types on protease activated receptors (PARs), metabotropic glutamate
of afferents tested, including esophageal, gastric, jejunal, colonic, lung, receptors (mGluRs) GABAB receptors and somatostatin receptors, as
carotid body, ureteric and urinary bladder afferents (Dang et al., 2008, well as mediators involved in satiety that act via vagal afferents.
2005; Cockayne et al., 2005; Wang & Neuhuber, 2003; Rong et al., 2002;
Brouns et al., 2000; Kirkup et al., 1999). 5. Protease activated receptors — PARs
In isolated visceral sensory neuronal cell bodies ATP and P2X
agonists activate inward currents (Xu et al., 2008; Dang et al., 2008). The protease activated receptors (PARs 1–4) are a recently described
The most important P2X subtypes in sensory neurons are thought to be class of G-protein coupled, seven transmembrane receptors that are
P2X2 and P2X3 which can form functional homomeric or heteromeric expressed in a variety of tissues (Coelho et al., 2003). The best
receptors. Thus experiments examining the role of P2X receptors in characterized on visceral afferents is PAR2. Its endogenous ligands
visceral sensory function have had to utilize double knockout animals. include tryptase and trypsin. Sources of these proteases include mast
In the urinary bladder, P2X2/3 double knockout animals had reduced cells and other inflammatory cells, digestive enzymes and possibly
ATP responses as well as reduced mechanosensation (Cockayne et al., bacteria themselves (Cottrell et al., 2003). In both nerve recording
2005). In contrast in the jejunum, mechanosensation was preserved in experiments and experiments on isolated cells, PAR-2 activation results
knockout mice, and was unaffected by P2X antagonism, but the in increased excitability, depolarization and afferent neuronal firing
enhanced excitability induced by inflammation was blunted by P2X (Kayssi et al., 2007; Kirkup et al., 2003) (see Fig. 3A). Recent studies have
antagonists, revealing a role for P2X 2/3 receptors in post inflammatory made significant progress in identifying the ionic mechanisms of action
states (Rong et al., 2009). of PAR-2, and its ion channel targets appear to be diverse. Firstly, PAR-2
is capable of inhibiting voltage gated potassium channels, in particular IK
4.3. Serotonin — 5HT3 currents, which are inhibited by more than 50% by PAR-2 activation
(Kayssi et al., 2007). There was no effect on voltage gated sodium
As greater than 90% of the body's 5-HT is found in the GI tract, it is currents, or A-type potassium currents (see Fig. 3B). These effects on
not surprising that there exists a serotonergic signaling system in excitability were dependent on the PKC and ERK pathways. In addition,
visceral afferents. 5HT3 receptors are known to be present on both PAR-2 is also capable of sensitizing both TRPV1 (Amadesi et al., 2006)
colonic (Hicks et al., 2002) and small intestinal afferent nerves and TRPV4 (Sipe et al., 2008) currents in colonic DRG neurons.
(Keating et al., 2008; Hillsley & Grundy, 1998; Hillsley et al., 1998; Li Furthermore, the ability of PAR-2 to evoke action potential discharge
et al., 2004). 5HT results in an increase in afferent firing in this from DRG neurons was significantly inhibited in mice deficient in
population that is nearly completely attenuated by selective antago- TRPV4, indicating an essential role in the actions of PAR-2, on visceral
nists of the 5-HT3 receptor. Furthermore in isolated abdominally afferents. Consistent with this, PAR-2 induced hyperalgesia was absent
projecting vagal afferent neurons, 5-HT results in inward currents in TRPV4 knockout animals as well (Sipe et al., 2008).
M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78 73

receptors enhance mechanosensitivity, suggesting the presence of


endogenous glutamatergic tone (Page et al., 2005). The source of
endogenous glutamate is not known, however afferent terminals
themselves do express glutamate transporters (Berthoud & Neuhuber,
2000) and glutamate may also be released from the enteric nervous
system (Wiley et al., 1991). Two separate reports suggest an
excitatory effect of a group I mGluR, mGluR5 on mechanosensitivity
in both gastroesophageal vagal afferents and colonic afferents (Page
et al., 2005; Lindstrom et al., 2008), with the latter paper providing
evidence for an effect on visceral pain. Interestingly, mGlur5 appears
to have a role as well in bladder afferent function, however at a
central, rather than peripheral level, as antagonism of this receptor
had no effect on bladder afferent discharge, but that systemic
administration inhibited mictruition. (Hu et al., 2009). Taken together
this line of work confirms the existence of both excitatory and
inhibitory mGluRs on gastrointestinal afferents. What remains to be
seen is how these competing influences interact in various patho-
physiological and physiological states. However they remain inter-
esting targets for drug targeting for disorders of GI motility and pain.

7. Somatostatin

Somatostatin is an endogenous peptide present in enteroendo-


crine D cells in the GI tract. Clinical studies suggest that somatostatin
agonists inhibit visceral sensation. A number of recent papers point to
a role for SST2 receptors in modulating visceral afferent excitability. In
the anaesthetized rat, SST2 agonists octreotide and BIM23027 have
effects on both vagal and spinal afferents (Booth et al., 2001). The
inhibitory effect on baseline discharge was inhibited by chronic
vagotomy, after which vagal afferent fibres projecting to the intestine
would be degenerated, suggesting a vagal site of action. However SST2
mediated inhibition of responses to distension and bradykinin were
not altered by vagotomy suggesting an effect on spinal afferents.
Subsequent experiments using animals deficient in the SST2 gene
revealed spontaneous hyperexcitability, and enhanced responses to
mechanical and chemical (acid) stimuli (Rong et al., 2007). This
suggests that endogenous somatostatin may act as a “brake” on the
excitability of intestinal afferents. Interestingly this system seems to
be downregulated in the post inflammatory state, and may contribute
to post inflammatory hyperexcitability (Aerssens et al., 2007).

8. GABAB

Gamma amino butyric acid type B receptors (GABAB) have also


been recently demonstrated to play a role in visceral afferent sen-
sitivity. Given the inhibitory effects of GABAB agonists on the transient
lower esophageal relaxations (TLESRs) that mediate gastroesophageal
reflux (Blackshaw et al., 2000; Partosoedarso et al., 2001), attention
has been directed to the afferent limb of this reflex, namely the vagal
afferent innervation of the proximal stomach and esophagus. GABAB
agonists inhibit mechanosensitive responses in vagal afferents
innervating the proximal stomach and esophagus (Page & Blackshaw,
1999) of the ferret. In the guinea pig, functional GABA receptors are
Fig. 3. PAR-2 excites visceral afferents by inhibiting potassium currents. A) PAR-2 agonists present in the nodose ganglion, however not in the afferent ending,
excite rat mesenteric afferents in vivo. Intravenous injection of SLIGRL, a PAR-2 agonist
indicating significant species differences with regard to this receptor.
results in an increase in mesenteric afferent firing. Whereas the reverse peptide LRGILS has
no effect (modified from Kirkup et al., 2003 with permission). B) Effect of PAR-2 on
The peripheral source of GABA is unknown. Nonetheless, GABAB re-
potassium currents in colon projecting DRG neurons. Application of the PAR-2 agonist mains an interesting target to reduce afferent input from the proximal
inhibits IK currents, but not IA (modified from Kayssi et al., 2007 with permission). stomach to reduce the incidence of TLESRs.

6. Metabotropic glutamate receptors 9. Satiety hormones

Recent work from the Blackshaw lab has pointed to a role for Given the emerging public heath problem of obesity, there is
metabotropic glutamate receptors in modulation of afferent sensitiv- increasing attention being focused on gastrointestinal derived satiety
ity. Receptor mRNA and protein for multiple mGluR subtypes are hormones and their action on afferents innervating the GI tract. Many
present in vagal afferents (Page et al., 2005). Agonists of group II and of these hormones exert satiety effects that at least partially depend
III receptors inhibit mechanosensitivity, and antagonists of these on gastrointestinal (vagal) afferents. Among the mediators studied,
74 M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78

glucagon like peptide-1 (GLP-1), cholecystokinin (CCK) and leptin endings (IGLEs) raise the tantalizing possibility of nitrergic crosstalk
have been directly shown to augment vagal afferent activity. In the between the enteric nervous system and extrinsic sensory afferents.
case of CCK, the receptor involved is known to be the CCK-1 (pre- With regard to H2S, there is an emerging yet conflicting body of
viously known as CCK-A) subtype. CCK increases firing in gastro- literature supporting its role in altering visceral afferent activity. The
intestinal afferent fibres innervating the stomach (Blackshaw & GI tract is unique in that H2S can both be derived from endogenous
Grundy, 1990), intestine (Li et al., 1999) and liver (Cox & Randich, sources, via the action of H2S producing enzyme systems, or from
1997) and activates isolated nodose ganglion cells (Peters et al., “exogenous sources” namely the product of bacterial metabolism by
2006b). Recent studies have also provided insight into some of the resident colonic bacteria (Moore et al., 2003). Initial reports suggested
potential ionic mechanisms of action. For example Ritter et al. have an antinociceptive role for H2S in the colon (Distrutti et al., 2006), as
shown that a nonselective cation conductance is activated by CCK in exogenous H2S inhibited visceromotor reflexes induced by colonic
nodose neurons, and potassium conductances are inhibited (Peters distention. These effects appeared to involve the activation of KATP
et al., 2006a). Preliminary work suggests that the cation conduc- channels, which would argue against involvement of primary af-
tance is pharmacologically similar to the TRPC channels (Beyak et al., ferents, as activation of potassium channels would tend to inhibit
2006b). Similarly the leptin receptor is expressed in human and afferent excitability. However a recent report suggests a pro-nociceptive
rodent nodose ganglion neurons, and leptin is known to activate role for luminal H2S, by evidence that pain related behaviours are
mesenteric afferents, as well as isolated nodose ganglion neurons enhanced by luminal H2S (Matsunami et al., 2009). The reasons for the
(Peters et al., 2006b; Buyse et al., 2001). Leptin appears to also both discrepancies in these findings are unclear, however possibilities
inhibit an outward current and activate an inward current, however include, secondary effects of H2S on smooth muscle tone, as H2S is a
the former action seems more prominent. known relaxer of GI smooth muscle (Gallego et al., 2008), or differences
With regard to GLP-1, there is emerging literature pointing to the in the concentrations of mediators involved. Importantly there are no
importance of visceral afferents of the vagus in its satiety and glucose published studies demonstrating the effects of H2S on visceral afferent
homeostatic effects (Abbott et al., 2005; Vahl et al., 2007), however, activity, however it appears to exert effects on bladder smooth muscle
the evidence demonstrating GLP-1 activating visceral afferents is still
sparse. There are direct electrophysiological recordings from hepatic
afferents showing an increase in firing rate in response to GLP-1
(Nishizawa et al., 2000), and nodose ganglia express the GLP-1
receptor mRNA (Nakagawa et al., 2004), and are depolarized by GLP-1
(Kakei et al., 2002). Preliminary work suggests an excitatory effect of
GLP-1 of intestinal afferents (Gaisano & Beyak, 2007), and a recent
report indicates that gastric vagal afferents are sensitive to GLP-1
in vivo (Bucinskaite et al., 2009). The mechanisms of action of GLP-1
are as yet not clear, however preliminary results from our lab suggest
an important role for inhibition of voltage gated potassium currents,
both IA and IK type currents (Gaisano & Beyak, 2007). PYY, another
distally released enteroendocrine hormone, appears to exert actions
on satiety and gastric emptying via vagal afferents however there are
no reports of it directly activating vagal afferents to date (Abbott et al.,
2005). On the topic of “orexigenic” i.e. appetite stimulating hormones,
the only ones to date studied are ghrelin and orexin. Ghrelin seems to
diminish vagal afferent firing (Date et al., 2002), and inhibit
mechanosensitivity (Page et al., 2007) which would tend to decrease
vagal satiety input to the brain. Orexin itself has no direct effect on
jejunal afferent activity, but inhibits the excitatory action of the satiety
hormone CCK (Burdyga et al., 2003).

10. Gasotransmitters NO/H2S

The original gaseous neurotransmitter described was nitric oxide


(NO) and in subsequent years both carbon monoxide (CO) and more
recently hydrogen sulfide (H2S) have been described. The literature for
these mediators affecting the discharge properties of afferent nerves
innervating the viscera is sparse, but has begun to emerge in recent
years.
A recent report suggests a role for peripheral nitric oxide in mod-
ulating the activity of gastrointestinal vagal afferent endings. Page
et al. (2009) have shown that neuronal nitric oxide synthase (nNOS)
inhibition enhanced mechanosensitivity, but only of those mechan-
osensitive mucosal afferents, not those presumed to be present in the
smooth muscle layer. In addition, exogenous NO donors inhibited Fig. 4. Endogenous nitric oxide inhibits visceral afferent mechanosensitivity. A) Inhibition
mechanosensitivity, as did exogenous stimulators of the cyclic GMP of endogenous NO production with L-NAME (open symbols) enhances responses of
pathway — the second messenger pathway acted upon by nitric oxide mucosal afferents to stroking with calibrated von Frey hairs. B) L-NAME has no effect on
(see Fig. 4). The source of endogenous NO mediating these actions responses on tension units (which respond to circumferential stretch). C) Raw nerve
tracing of a mucosal unit and its responses to stroking of the receptive field with 50 mg von
is thought to be mucosal enteroendocrine cells expressing nNOS, Frey hairs. i) control trace ii) note enhancement of responses after L-NAME iii) L-arginine,
though the presence of specialized afferent endings terminating in the the natural substrate of nitric oxide synthase reverses the effects of L-NAME (modified
myenteric ganglia themselves, the so called intraganglionic laminar from Page et al., 2009 with permission).
M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78 75

contraction and intestinal secretion via an action on capsaicin sensitive inflammatory changes had resolved. Recordings from colonic afferent
visceral afferents (Patacchini et al., 2004; Schicho et al., 2006). There is nerve fibres after the resolution of inflammation also revealed increased
however one preliminary study demonstrating an excitatory effect of mechanosensitive responses (Ibeakanma et al., 2009). Recent papers
H2S on mesenteric afferent firing (Rong et al., 5 A.D.). Studies of the have also identified that after resolution of inflammation induced by
direct effects of hydrogen sulfide on afferent excitability will be critical TNBS, a model of inflammatory bowel disease, that hyperexcitiability of
in unraveling this potentially important novel signaling pathway. colonic afferents is present both during acute inflammation and after
recovery, though hypersensitivity was only observed in the acute phase
11. Chronic alteration in visceral excitability in splanchnic afferents, and not pelvic fibres, suggesting differences
between subtypes of fibres. (Hughes et al., 2009)
The above sections have focused on mechanisms by which the
firing properties of visceral afferents can be altered on an “acute” or
minute to minute basis. However there are clearly situations where 11.2. Mechanisms of inflammation induced hyperexcitability — alterations
the function of these nerves is altered on a more long term basis, in receptor and ion channel function
causing relative “hypersensitivity” or “hyposensitivity.” These con-
cepts are relevant in disease states such as visceral pain, where normal Inflammation clearly causes alteration in the membrane properties
visceral functions often result in painful sensations (visceral allody- of visceral afferent neurons, and this suggests a fundamental alter-
nia) or exaggerated responses to normally painful stimuli (visceral ation in the major determinants of neuronal excitability, namely vol-
hyperalgesia). Indeed observations of altered sensory thresholds exist tage gated potassium and sodium channels. With regard to voltage
in a variety of visceral pain disorders such as noncardiac chest pain, gated potassium currents a variety of models of acute inflammation in
functional dyspepsia, irritable bowel syndrome and interstitial
cystitis. With this in mind, many investigators have developed models
of visceral disease, typically inflammation, to study the medium and
long term impact on the function of visceral afferents. The review by
Vergnolle in this issue will discuss in detail numerous aspects of the
effects of inflammation on visceral afferent function.

11.1. Inflammation and afferent excitability

As it is intuitive that inflammation results in pain, it follows that a


number of models of inflammation have been used to study the
impact of visceral afferent function. Studies using recordings from
isolated sensory neurons from the dorsal root ganglion have provided
a number of insights. Yoshimura and DeGroat first characterized
changes in excitability of bladder projecting DRG neurons in a model
of cyclophosphamide induced cystitis (Yoshimura & de Groat, 1999).
Neurons projecting to the inflamed bladder (identified by retrograde
labeling) exhibited lower current thresholds, fired more action
potentials at suprathreshold stimulus strengths, and had greater
input resistances. Subsequently studies using a model of inflamma-
tory bowel disease (trinitrobenzene sulphonic acid — TNBS) in the
ileum (Stewart et al., 2003) and colon (Beyak et al., 2004), and models
of ulcer disease (Dang et al., 2004) in the stomach have also revealed
similar effects on current threshold (rheobase) and repetitive firing. It
is important that these experiments were performed on cells removed
from the inflammatory in vivo milieu for several hours, suggesting the
presence of longer term changes in excitability, eliminating the types
of influences discussed in the previous sections.
The above important studies examined the effects of active inflam-
mation, but as many human conditions of visceral hypersensitivity
such as IBS occur in the absence of obvious inflammation, the question
remained whether these changes can persist after the resolution of
inflammation. Two recent studies have addressed this issue using
differing models of self limited gastrointestinal infection. Keating et al.
demonstrated that after resolution of the acute inflammatory phase of
intestinal infection with Trichinella spiralis that there were increased
responses to mechanical and chemical stimuli at the level of the afferent
terminal, and that hyperexcitability was present in isolated labeled gut
projecting sensory neurons at this time point as well — similar to
changes previously seen on neurons recorded from during the acute
Fig. 5. Increased excitability of visceral afferents in the post inflammatory state.
phase of inflammation. (Keating et al., 2008) (see Fig. 5) A more recent Transient inflammation was induced with infection with Trichinella spiralis. A) jejunal
study by Ibeakanma et al. used a mouse model of Citrobacter rodentium afferent responses to distension in the control, acute inflammatory period (day 14–16
infection, a self limited bacterial infectious colitis, similar to that seen post infection) and post inflammatory periods (after resolution of inflammation).
with pathogenic E. coli infection in humans. Again similar changes of During acute inflammation, there is a reduction in mechanosensitive responses,
however after resolution, there is evidence of increased response of jejunal afferent
decreased rheobase, and increased repetitive firing were seen in the nerves to distention. B) In isolated nodose ganglion neurons, there is evidence of
acute inflammatory period, and the increased repetitive DRG neuronal decreased rheobase and increased repetitive firing at twice rheobase in the post
firing persisted in post infections state, at time points when the acute inflammatory period (modified from Keating et al., 2008 with permission).
76 M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78

the stomach (Dang et al., 2004), ileum (Stewart et al., 2003), colon
(Ibeakanma et al., 2009) and urinary bladder (Yoshimura & de Groat,
1999) have suggested that voltage gated currents are suppressed, and
their steady state inactivation properties altered such that fewer
channels are available for activation. Indeed the effects of TNBS ileitis
on labeled DRG neurons was very similar to that of administration of
the IA blocker 4-aminopyridine (Moore et al., 2002). Whether or not
only IA or IA and IK type currents are suppressed vary between
studies; however in most studies the suppression of IA is a consistent
finding. Following the resolution of inflammation, these suppressive
effects on potassium currents, at least IA, seem to persist (Ibeakanma
et al., 2009). Fig. 6. Enhanced P2X receptor function in bladder projecting DRG neurons following
Important changes in voltage gated sodium channels have also been inflammation. Left panels are currents evoked by superfusion of ATP, right panels are
currents evoked by superfusion of the P2X agonist αβ-methylene ATP. B represents
demonstrated in inflammation. In TNBS colitis and ileitis, TTX resistant currents from control neurons, B⁎ represents currents in neurons from animals with
sodium currents are increased, and their activation thresholds reduced cystitis. After inflammation currents are significantly increased (modified from Dang
(Beyak et al., 2004; Stewart et al., 2003). Other investigators examining et al., 2008 with permission).
bladder, and stomach afferents (Bielefeldt et al., 2002a,b) have made
similar findings. The effect on NaV currents is relatively selective. In 12. Visceral hyposensitivity
addition the kinetics of the channels are also altered, with the speed of
recovery increasing after inflammation, and the threshold for activation While the investigation of the pathogenesis of visceral pain has
is lowered (Beyak et al., 2004). The increase in this rapidly repriming driven the study of visceral afferent hypersensitivity, the topic of
TTX-R current, combined with the decreased activation threshold would visceral hyposensitivity has been relatively neglected by all but a few
tend to result in increased ability to fire repetitively with a lower researchers. In humans hyposensitivity to visceral stimuli may
threshold for action potential electrogenesis. The critical role for the NaV underlie constipation and defacatory problems. (Gladman et al.,
1.8 TTX-R current is illustrated using elegant studies by Hillsley et al. 2009) Certainly there is impairment of somatic and probably visceral
(2006), in which transient jejunitis due to infection with Nippostron- afferent function in disorders such as diabetes mellitus. In incontinent
gylus brasiliensis induced hyperexcitability of intestine projecting DRG patients with diabetes, there is significantly impaired detection of
neurons, and increased TTX-R currents. These investigators used NaV 1.8 normal rectal filling (Caruana et al., 1991; Sun et al., 1996; Wald &
knockout mice, and showed that post infectious hyperexcitability was Tunuguntla, 1984). A recent paper examining rectal afferent function
absent in these animals. Whether the observed changes in TTX-R in a rat model of diabetes suggests that there is significant impairment
currents are due to alterations in channel expression or to post- of fibres that detect low threshold stretch, with relative preservation
translational modeling is an important question. A recent paper has of responses to near noxious levels of mechanical stimulation (Beyak
attempted to address this issue, examining protein expression of im- et al., 2009). The mechanisms for this are unclear, and this is an open
portant channel subunits NaV 1.7, 1.8 and 1.9 with western blot tech- field for investigation. In addition there is ample evidence for
niques as well as examining mRNA levels using quantitative rtPCR impaired satiety related afferent input to the CNS in diet induced
performed on labeled colon projecting neurons by laser capture micro- obesity in humans and animals (Covasa et al., 2000a,b), however there
dissection in animals with and without TNBS colitis. At time points has been little to no direct investigation of responses of satiety related
corresponding to previous studies showing enhanced excitability, vagal afferents in models of obesity. Perhaps the impaired within meal
expression of NaV1.8 protein was increased, however there was no feedback signaling that appears to be present in both obese animals
change in mRNA, suggesting that changes are occurring at the post- and humans could be due to hyposensitivity of satiety related vagal
translational level (King et al., 2009). afferents innervating the stomach, liver and intestine. However this
In addition to the aforementioned changes in voltage gated hypothesis remains to be tested.
channels, there is also evidence for changes in the number and or
function of ligand gated channels as well. With regard to TRPV1, a 13. Summary and conclusions
variety of models of inflammation in the viscera up regulate its
expression, and in a number of models is necessary for pain related Visceral afferents display a wide range of responsiveness to
behaviours as well as afferent nerve mechanohypersensitivity (De mechanical and chemical stimuli. The basic determinants of excit-
Schepper et al., 2008; Phillis et al., 2009). In addition, a recent ability are the membrane voltage gated sodium and potassium
interesting report suggested that down regulation of TRPA1 atten- channels, and the multitudes of mediators that act on them impact
uates colitis induced hyperalgesia (Yang et al., 2008). P2X currents the function of these channels directly or indirectly. Furthermore the
also seem to be unregulated by gastric (Dang et al., 2005) and ability to change in the long term the properties of the determinants
bladder (Dang et al., 2008) (see Fig. 6) inflammations as are afferent of excitability results in states of hyper- and possibly hypo-excitability
neural responses to ATP. The situation with 5-HT may however be and have important implications for a variety of disease states such as
different. Keating et al. (2008) showed that despite increased visceral pain, diabetes and obesity.
responses to exogenous 5-HT, and increased 5-HT availability, that
5-HT mediated currents were actually decreased in the post inflam- References
matory state. This suggested that in some instances, receptors may
Abbott, C.R., Monteiro, M., Small, C.J., Sajedi, A., Smith, K.L., Parkinson, J.R., Ghatei, M.A.,
down regulate in the face of increased availability of the ligand.
Bloom, S.R., 2005. The inhibitory effects of peripheral administration of peptide YY
Taken together, the evidence to date is that visceral inflammation (3–36) and glucagon-like peptide-1 on food intake are attenuated by ablation of
results in multiple changes in ion channel function, including aug- the vagal–brainstem–hypothalamic pathway. Brain Res. 1044, 127–131.
Aerssens, J., Hillsley, K., Peeters, P.J., de, H.R., Stanisz, A., Lin, J.H., Van dW, I., Gohlmann,
menting of sodium channel function, while at the same time inhibiting
H.W., Grundy, D., Stead, R.H., Coulie, B., 2007. Alterations in the brain-gut axis
potassium channels that normally allow a neuron to accommodate a underlying visceral chemosensitivity in Nippostrongylus brasiliensis-infected mice.
depolarizing stimulus, thus leading to decreased action potential Gastroenterology 132, 1375–1387.
thresholds and repetitive firing. The enhancement of function of a Amadesi, S., Cottrell, G.S., Divino, L., Chapman, K., Grady, E.F., Bautista, F., Karanjia, R.,
Barajas-Lopez, C., Vanner, S., Vergnolle, N., Bunnett, N.W., 2006. Protease-activated
number of ligand gated channels further increases the effect that ligands receptor 2 sensitizes TRPV1 by protein kinase Cepsilon- and A-dependent
will have on the nerve terminal. mechanisms in rats and mice. J. Physiol. 575, 555–571.
M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78 77

Berthoud, H.R., Neuhuber, W.L., 2000. Functional and chemical anatomy of the afferent Date, Y., Murakami, N., Toshinai, K., Matsukura, S., Niijima, A., Matsuo, H., Kangawa, K.,
vagal system. Auton. Neurosci. 85, 1–17. Nakazato, M., 2002. The role of the gastric afferent vagal nerve in ghrelin-induced
Beyak, M.J., Ramji, N., Krol, K.M., Kawaja, M.D., Vanner, S.J., 2004. Two TTX-resistant feeding and growth hormone secretion in rats. Gastroenterology 123, 1120–1128.
Na+ currents in mouse colonic dorsal root ganglia neurons and their role in De Schepper, H.U., De Winter, B.Y., Van, N.L., Timmermans, J.P., Herman, A.G.,
colitis-induced hyperexcitability. Am. J. Physiol.: Gastrointest. Liver Physiol. 287, Pelckmans, P.A., De Man, J.G., 2008. TRPV1 receptors on unmyelinated C-fibres
G845–G855. mediate colitis-induced sensitization of pelvic afferent nerve fibres in rats.
Beyak, M., Bulmer, D.C., Jiang, W., Keating, C., Rong, W., Grundy, D., 2006a. Sensory afferent J. Physiol. 586, 5247–5258.
nerves innervating the gastrointestinal tract. In: Johnson, LR (Ed.), Physiology of the Distrutti, E., Sediari, L., Mencarelli, A., Renga, B., Orlandi, S., Antonelli, E., Roviezzo, F.,
Gastrointestinal Tract. Elsevier Press, New York. Morelli, A., Cirino, G., Wallace, J.L., Fiorucci, S., 2006. Evidence that hydrogen sulfide
Beyak, M.J., Surprenant, A., Grundy, D., 2006b. Stimulation of CCK-1 receptors activates exerts antinociceptive effects in the gastrointestinal tract by activating KATP
a TRPC like inward current in GI vagal afferent neurons. Gastroenterology 130 (4), channels. J. Pharmacol. Exp. Ther. 316, 325–335.
A74 (Ref Type: Abstract). Fang, X., Djouhri, L., McMullan, S., Berry, C., Waxman, S.G., Okuse, K., Lawson, S.N.,
Beyak, M.J., Bulmer, D.C., Sellers, D., Grundy, D., 2009. Impairment of rectal afferent 2006. Intense isolectin-B4 binding in rat dorsal root ganglion neurons distin-
mechanosensitivity in experimental diabetes in the rat. Neurogastroenterol. Motil. guishes C-fiber nociceptors with broad action potentials and high Nav1.9
21 (6), 678–681. expression. J. Neurosci. 26, 7281–7292.
Bielefeldt, K., Davis, B.M., 2008. Differential effects of ASIC3 and TRPV1 deletion on Gaisano, G., Beyak, M., 2007. The GLP-1 agonist exendin-4 excites nodose ganglion neurons
gastroesophageal sensation in mice. Am. J. Physiol.: Gastrointest. Liver Physiol. and inhibits voltage-gated potassium currents. Gastroenterology 132 (4 (suppl2)),
294, G130–G138. A36 (Ref Type: Abstract).
Bielefeldt, K., Ozaki, N., Gebhart, G.F., 2002a. Experimental ulcers alter voltage- Gallego, D., Clave, P., Donovan, J., Rahmati, R., Grundy, D., Jimenez, M., Beyak, M.J., 2008. The
sensitive sodium currents in rat gastric sensory neurons. Gastroenterology 122, gaseous mediator, hydrogen sulphide, inhibits in vitro motor patterns in the human,
394–405. rat and mouse colon and jejunum. Neurogastroenterol. Motil. 20, 1306–1316.
Bielefeldt, K., Ozaki, N., Gebhart, G.F., 2002b. Mild gastritis alters voltage-sensitive Gladman, M.A., Aziz, Q., Scott, S.M., Williams, N.S., Lunniss, P.J., 2009. Rectal hyposensi-
sodium currents in gastric sensory neurons in rats. Gastroenterology 122, 752–761. tivity: pathophysiological mechanisms. Neurogastroenterol. Motil. 21, 508-5.
Black, J.A., Cummins, T.R., Yoshimura, N., de Groat, W.C., Waxman, S.G., 2003. Hay, M., Kunze, D.L., 1994a. An intermediate conductance calcium-activated potassium
Tetrodotoxin-resistant sodium channels Na (v)1.8/SNS and Na (v)1.9/NaN in afferent channel in rat visceral sensory afferent neurons. Neurosci. Lett. 167, 179–182.
neurons innervating urinary bladder in control and spinal cord injured rats. Brain Res. Hay, M., Kunze, D.L., 1994b. Calcium-activated potassium channels in rat visceral
963, 132–138. sensory afferents. Brain Res. 639, 333–336.
Blackshaw, L.A., Grundy, D., 1990. Effects of cholecystokinin (CCK-8) on two classes of Hicks, G.A., Coldwell, J.R., Schindler, M., Ward, P.A., Jenkins, D., Lynn, P.A., Humphrey, P.P.,
gastroduodenal vagal afferent fibre. J. Auton. Nerv. Syst. 31, 191–201. Blackshaw, L.A., 2002. Excitation of rat colonic afferent fibres by 5-HT (3) receptors.
Blackshaw, L.A., Gebhart, G.F., 2002. The pharmacology of gastrointestinal nociceptive J. Physiol. 544, 861–869.
pathways. Curr. Opin. Pharmacol. 2, 642–649. Hille, B., 2001. Classical biophysics of the squid giant axon. Ion Channels of Excitable
Blackshaw, L.A., Brookes, S.J., Grundy, D., Schemann, M., 2007. Sensory transmission in Membranes. Sinauer Associates, Sunderland, pp. 25–59.
the gastrointestinal tract. Neurogastroenterol. Motil. 19, 1–19. Hillsley, K., Grundy, D., 1998. Sensitivity to 5-hydroxytryptamine in different afferent
Blackshaw, L.A., Smid, S.D., O'Donnell, T.A., Dent, J., 2000. GABA (B) receptor-mediated subpopulations within mesenteric nerves supplying the rat jejunum. J. Physiol. 509
effects on vagal pathways to the lower oesophageal sphincter and heart. Br. J. (Pt 3), 717–727.
Pharmacol. 130, 279–288. Hillsley, K., Kirkup, A.J., Grundy, D., 1998. Direct and indirect actions of 5-hydroxytrypta-
Booth, C.E., Kirkup, A.J., Hicks, G.A., Humphrey, P.P., Grundy, D., 2001. Somatostatin sst mine on the discharge of mesenteric afferent fibres innervating the rat jejunum. J.
(2) receptor-mediated inhibition of mesenteric afferent nerves of the jejunum in Physiol. 506 (Pt 2), 551–561.
the anesthetized rat. Gastroenterology 121, 358–369. Hillsley, K., Lin, J.H., Stanisz, A., Grundy, D., Aerssens, J., Peeters, P.J., Moechars, D., Coulie,
Brierley, S.M., Page, A.J., Hughes, P.A., Adam, B., Liebregts, T., Cooper, N.J., Holtmann, G., B., Stead, R.H., 2006. Dissecting the role of sodium currents in visceral sensory
Liedtke, W., Blackshaw, L.A., 2008. Selective role for TRPV4 ion channels in visceral neurons in a model of chronic hyperexcitability using Nav1.8 and Nav1.9 null mice.
sensory pathways. Gastroenterology 134, 2059–2069. J. Physiol. 576, 257–267.
Brouns, I., Adriaensen, D., Burnstock, G., Timmermans, J.P., 2000. Intraepithelial vagal Hu, Y., Dong, L., Sun, B., Guillon, M.A., Burbach, L.R., Nunn, P.A., Liu, X., Vilenski, O., Ford,
sensory nerve terminals in rat pulmonary neuroepithelial bodies express P2X (3) A.P., Zhong, Y., Rong, W., 2009. The role of metabotropic glutamate receptor mGlu5
receptors. Am. J. Respir. Cell Mol. Biol. 23, 52–61. in control of micturition and bladder nociception. Neurosci. Lett. 450, 12–17.
Bucinskaite, V., Tolessa, T., Pedersen, J., Rydqvist, B., Zerihun, L., Holst, J.J., Hellstrom, P.M., Hughes, P.A., Brierley, S.M., Martin, C.M., Brookes, S.J., Linden, D.R., Blackshaw, L.A.,
2009. Receptor-mediated activation of gastric vagal afferents by glucagon-like- 2009. Post-inflammatory colonic afferent sensitization: different subtypes, differ-
peptide-1 in the rat. Neurogastroenterol. Motil. 21 (9), 978–984. ent pathways, and different time-courses. Gut. 58, 1333–1431.
Burdyga, G., Lal, S., Spiller, D., Jiang, W., Thompson, D., Attwood, S., Saeed, S., Grundy, D., Ibeakanma, C.O., Miranda-Morales, M., Richards, M., Bautista-Cruz, F., Martin, N.,
Varro, A., Dimaline, R., Dockray, G.J., 2003. Localization of orexin-1 receptors to Hurlbut, D., Vanner, S., 2009. Citrobacter rodentium colitis evokes post-infectious
vagal afferent neurons in the rat and humans. Gastroenterology 124, 129–139. hyperexcitability of mouse nociceptive colonic DRG neurons. J. Physiol. 587,
Buyse, M., Ovesjo, M.L., Goiot, H., Guilmeau, S., Peranzi, G., Moizo, L., Walker, F., Lewin, M.J., 3505–3521.
Meister, B., Bado, A., 2001. Expression and regulation of leptin receptor proteins in Jones, R.C., Xu, L., Gebhart, G.F., 2005. The mechanosensitivity of mouse colon afferent
afferent and efferent neurons of the vagus nerve. Eur. J. Neurosci. 14, 64–72. fibers and their sensitization by inflammatory mediators require transient receptor
Caruana, B.J., Wald, A., Hinds, J.P., Eidelman, B.H., 1991. Anorectal sensory and motor potential vanilloid 1 and acid-sensing ion channel 3. J. Neurosci. 25, 10981–10989.
function in neurogenic fecal incontinence. Comparison between multiple sclerosis Kakei, M., Yada, T., Nakagawa, A., Nakabayashi, H., 2002. Glucagon-like peptide-1 evokes
and diabetes mellitus. Gastroenterology 100, 465–470. action potentials and increases cytosolic Ca2+ in rat nodose ganglion neurons.
Castelucci, P., Robbins, H.L., Furness, J.B., 2003. P2X (2) purine receptor immunor- Auton. Neurosci. 102, 39–44.
eactivity of intraganglionic laminar endings in the mouse gastrointestinal tract. Cell Kayssi, A., Amadesi, S., Bautista, F., Bunnett, N.W., Vanner, S., 2007. Mechanisms of
Tissue Res. 312, 167–174. protease-activated receptor 2-evoked hyperexcitability of nociceptive neurons
Cockayne, D.A., Dunn, P.M., Zhong, Y., Rong, W., Hamilton, S.G., Knight, G.E., Ruan, H.Z., Ma, innervating the mouse colon. J. Physiol. 580, 977–991.
B., Yip, P., Nunn, P., McMahon, S.B., Burnstock, G., Ford, A.P., 2005. P2X2 knockout mice Keating, C., Beyak, M., Foley, S., Singh, G., Marsden, C., Spiller, R., Grundy, D., 2008.
and P2X2/P2X3 double knockout mice reveal a role for the P2X2 receptor subunit in Afferent hypersensitivity in a mouse model of post-inflammatory gut dysfunction:
mediating multiple sensory effects of ATP. J. Physiol. 567, 621–639. role of altered serotonin metabolism. J. Physiol. 586, 4517–4530.
Coelho, A.M., Ossovskaya, V., Bunnett, N.W., 2003. Proteinase-activated receptor-2: Kim, D.S., Choi, J.O., Rim, H.D., Cho, H.J., 2002. Downregulation of voltage-gated po-
physiological and pathophysiological roles. Curr. Med. Chem. Cardiovasc. Hematol. tassium channel alpha gene expression in dorsal root ganglia following chronic
Agents 1, 61–72. constriction injury of the rat sciatic nerve. Brain Res. Mol. Brain Res. 105, 146–152.
Cottrell, G.S., Amadesi, S., Schmidlin, F., Bunnett, N., 2003. Protease-activated receptor King, D.E., Macleod, R.J., Vanner, S.J., 2009. Trinitrobenzenesulphonic acid colitis alters Na
2: activation, signalling and function. Biochem. Soc. Trans. 31, 1191–1197. 1.8 channel expression in mouse dorsal root ganglia neurons. Neurogastroenterol.
Covasa, M., Grahn, J., Ritter, R.C., 2000a. High fat maintenance diet attenuates hindbrain Motil. 21 (8), 880–e64.
neuronal response to CCK. Regul. Pept. 86, 83–88. Kirkup, A.J., Booth, C.E., Chessell, I.P., Humphrey, P.P., Grundy, D., 1999. Excitatory effect
Covasa, M., Grahn, J., Ritter, R.C., 2000b. Reduced hindbrain and enteric neuronal response of P2X receptor activation on mesenteric afferent nerves in the anaesthetised rat.
to intestinal oleate in rats maintained on high-fat diet. Auton. Neurosci. 84, 8–18. J. Physiol. 520 (Pt. 2), 551–563.
Cox, J.E., Randich, A., 1997. CCK-8 activates hepatic vagal C-fiber afferents. Brain Res. Kirkup, A.J., Brunsden, A.M., Grundy, D., 2001. Receptors and transmission in the brain-gut
776, 189–194. axis: potential for novel therapies. I. Receptors on visceral afferents. Am. J. Physiol.:
Daly, D., Rong, W., Chess-Williams, R., Chapple, C., Grundy, D., 2007. Bladder afferent Gastrointest. Liver Physiol. 280, G787–G794.
sensitivity in wild-type and TRPV1 knockout mice. J. Physiol. 583, 663–674. Kirkup, A.J., Jiang, W., Bunnett, N.W., Grundy, D., 2003. Stimulation of proteinase-
Dang, K., Bielefeldt, K., Gebhart, G.F., 2004. Gastric ulcers reduce A-type potassium activated receptor 2 excites jejunal afferent nerves in anaesthetised rats. J. Physiol.
currents in rat gastric sensory ganglion neurons. Am. J. Physiol.: Gastrointest. Liver 552, 589–601.
Physiol. 286, G573–G579. Li, Y., Zhu, J., Owyang, C., 1999. Electrical physiological evidence for highand low-
Dang, K., Bielfeldt, K., Lamb, K., Gebhart, G.F., 2005. Gastric ulcers evoke hyperexcitability affinity vagal CCK-A receptors. Am. J. Physiol. 277, G469–G477.
and enhance P2X receptor function in rat gastric sensory neurons. J. Neurophysiol. Li, Y., Wu, X.Y., Owyang, C., 2004. Serotonin and cholecystokinin synergistically
93, 3112–3119. stimulate rat vagal primary afferent neurones. J. Physiol. 559, 651–662.
Dang, K., Lamb, K., Cohen, M., Bielefeldt, K., Gebhart, G.F., 2008. Cyclophosphamide- Lindstrom, E., Brusberg, M., Hughes, P.A., Martin, C.M., Brierley, S.M., Phillis, B.D.,
induced bladder inflammation sensitizes and enhances P2X receptor function in rat Martinsson, R., Abrahamsson, C., Larsson, H., Martinez, V., Blackshaw, L.A., 2008.
bladder sensory neurons. J. Neurophysiol. 99, 49–59. Involvement of metabotropic glutamate 5 receptor in visceral pain. Pain 137, 295–305.
78 M.J. Beyak / Autonomic Neuroscience: Basic and Clinical 153 (2010) 69–78

Matsunami, M., Tarui, T., Mitani, K., Nagasawa, K., Fukushima, O., Okubo, K., Yoshida, S., Rong, W., Keating, C., Sun, B., Dong, L., Grundy, D., 2009. Purinergic contribution to small
Takemura, M., Kawabata, A., 2009. Luminal hydrogen sulfide plays a pronociceptive intestinal afferent hypersensitivity in a murine model of postinfectious bowel disease.
role in mouse colon. Gut 58, 751–761. Neurogastroenterol. Motil. 21 (6), 665–e32.
McIlwrath, S.L., Davis, B.M., Bielefeldt, K., 2009. Deletion of P2X3 receptors blunts Schicho, R., Krueger, D., Zeller, F., Von Weyhern, C.W., Frieling, T., Kimura, H., Ishii, I., De, G.R.,
gastro-oesophageal sensation in mice. Neurogastroenterol. Motil. 21 (8), 890–e66. Campi, B., Schemann, M., 2006. Hydrogen sulfide is a novel prosecretory neuromo-
Moore, B.A., Stewart, T.M., Hill, C., Vanner, S.J., 2002. TNBS ileitis evokes hyperexcit- dulator in the guinea-pig and human colon. Gastroenterology 131, 1542–1552.
ability and changes in ionic membrane properties of nociceptive DRG neurons. Am. Sipe, W.E., Brierley, S.M., Martin, C.M., Phillis, B.D., Cruz, F.B., Grady, E.F., Liedtke, W.,
J. Physiol.: Gastrointest. Liver Physiol. 282, G1045–G1051. Cohen, D.M., Vanner, S., Blackshaw, L.A., Bunnett, N.W., 2008. Transient receptor
Moore, P.K., Bhatia, M., Moochhala, S., 2003. Hydrogen sulfide: from the smell of the potential vanilloid 4 mediates protease activated receptor 2-induced sensitization
past to the mediator of the future? Trends Pharmacol. Sci. 24, 609–611. of colonic afferent nerves and visceral hyperalgesia. Am. J. Physiol.: Gastrointest.
Nakagawa, A., Satake, H., Nakabayashi, H., Nishizawa, M., Furuya, K., Nakano, S., Kigoshi, T., Liver Physiol. 294, G1288–G1298.
Nakayama, K., Uchida, K., 2004. Receptor gene expression of glucagon-like peptide-1, Stewart, T., Beyak, M.J., Vanner, S., 2003. Ileitis modulates potassium and sodium
but not glucose-dependent insulinotropic polypeptide, in rat nodose ganglion cells. currents in guinea pig dorsal root ganglia sensory neurons. J. Physiol. 552, 797–807.
Auton. Neurosci. 110, 36–43. Sugiuar, T., Bielefeldt, K., Gebhart, G.F., 2004. TRPV1 function in mouse colon sensory
Nishizawa, M., Nakabayashi, H., Kawai, K., Ito, T., Kawakami, S., Nakagawa, A., Niijima, A., neurons is enhanced by metabotropic 5-hydroxytryptamine receptor activation.
Uchida, K., 2000. The hepatic vagal reception of intraportal GLP-1 is via receptor J. Neurosci. 24, 9521–9530.
different from the pancreatic GLP-1 receptor. J. Auton. Nerv. Syst. 80, 14–21. Sugiura, T., Bielefeldt, K., Gebhart, G.F., 2007. Mouse colon sensory neurons detect
Page, A.J., Blackshaw, L.A., 1999. GABA (B) receptors inhibit mechanosensitivity of extracellular acidosis via TRPV1. Am. J. Physiol., Cell Physiol. 292, C1768–C1774.
primary afferent endings. J. Neurosci. 19, 8597–8602. Sun, W.M., Katsinelos, P., Horowitz, M., Read, N.W., 1996. Disturbances in anorectal function
Page, A.J., Young, R.L., Martin, C.M., Umaerus, M., O'Donnell, T.A., Cooper, N.J., Coldwell, in patients with diabetes mellitus and faecal incontinence. Eur. J. Gastroenterol.
J.R., Hulander, M., Mattsson, J.P., Lehmann, A., Blackshaw, L.A., 2005. Metabotropic Hepatol. 8, 1007–1012.
glutamate receptors inhibit mechanosensitivity in vagal sensory neurons. Gastro- Vahl, T.P., Tauchi, M., Durler, T.S., Elfers, E.E., Fernandes, T.M., Bitner, R.D., Ellis, K.S., Woods,
enterology 128, 402–410. S.C., Seeley, R.J., Herman, J.P., D'Alessio, D.A., 2007. Glucagon-like peptide-1 (GLP-1)
Page, A.J., Slattery, J.A., Milte, C., Laker, R., O'Donnell, T., Dorian, C., Brierley, S.M., Blackshaw, receptors expressed on nerve terminals in the portal vein mediate the effects of
L.A., 2007. Ghrelin selectively reduces mechanosensitivity of upper gastrointestinal endogenous GLP-1 on glucose tolerance in rats. Endocrinology 148, 4965–4973.
vagal afferents. Am. J. Physiol.: Gastrointest. Liver Physiol. 292, G1376–G1384. Wald, A., Tunuguntla, A.K., 1984. Anorectal sensorimotor dysfunction in fecal incon-
Page, A.J., O'Donnell, T.A., Cooper, N.J., Young, R.L., Blackshaw, L.A., 2009. Nitric oxide as an tinence and diabetes mellitus. Modification with biofeedback therapy. N. Engl. J.
endogenous peripheral modulator of visceral sensory neuronal function. J. Neurosci. Med. 310, 1282–1287.
29, 7246–7255. Wang, Z.J., Neuhuber, W.L., 2003. Intraganglionic laminar endings in the rat esophagus
Park, S.Y., Choi, J.Y., Kim, R.U., Lee, Y.S., Cho, H.J., Kim, D.S., 2003. Downregulation of contain purinergic P2X2 and P2X3 receptor immunoreactivity. Anat. Embryol.
voltage-gated potassium channel alpha gene expression by axotomy and (Berl) 207, 363–371.
neurotrophins in rat dorsal root ganglia. Mol. Cells 16, 256–259. Waxman, S.G., Cummins, T.R., Dib-Hajj, S., Fjell, J., Black, J.A., 1999a. Sodium channels,
Partosoedarso, E.R., Young, R.L., Blackshaw, L.A., 2001. GABA (B) receptors on vagal excitability of primary sensory neurons, and the molecular basis of pain. Muscle
afferent pathways: peripheral and central inhibition. Am. J. Physiol.: Gastrointest. Nerve 22, 1177–1187.
Liver Physiol. 280, G658–G668. Waxman, S.G., Cummins, T.R., Dib-Hajj, S., Fjell, J., Black, J.A., 1999b. Sodium channels,
Patacchini, R., Santicioli, P., Giuliani, S., Maggi, C.A., 2004. Hydrogen sulfide (H2S) excitability of primary sensory neurons, and the molecular basis of pain. Muscle
stimulates capsaicin-sensitive primary afferent neurons in the rat urinary bladder. Nerve 22, 1177–1187.
Br. J. Pharmacol. 142, 31–34. Wiley, J.W., Lu, Y.X., Owyang, C., 1991. Evidence for a glutamatergic neural pathway in
Patapoutian, A., Tate, S., Woolf, C.J., 2009. Transient receptor potential channels: the myenteric plexus. Am. J. Physiol. 261, G693–G700.
targeting pain at the source. Nat. Rev. Drug Discov. 8, 55–68. Xu, G.Y., Shenoy, M., Winston, J.H., Mittal, S., Pasricha, P.J., 2008. P2X receptor-
Peters, J.H., Ritter, R.C., Simasko, S.M., 2006a. Leptin and CCK modulate complementary mediated visceral hyperalgesia in a rat model of chronic visceral hypersensitivity.
background conductances to depolarize cultured nodose neurons. Am. J. Physiol., Gut 57, 1230–1237.
Cell Physiol. 290, C427–C432. Yang, E.K., Takimoto, K., Hayashi, Y., de Groat, W.C., Yoshimura, N., 2004. Altered
Peters, J.H., Ritter, R.C., Simasko, S.M., 2006b. Leptin and CCK selectively activate vagal expression of potassium channel subunit mRNA and alpha-dendrotoxin sensitivity of
afferent neurons innervating the stomach and duodenum. Am. J. Physiol., Regul. potassium currents in rat dorsal root ganglion neurons after axotomy. Neuroscience
Integr. Comp. Physiol. 290, R1544–R1549. 123, 867–874.
Phillis, B.D., Martin, C.M., Kang, D., Larsson, H., Lindstrom, E.A., Martinez, V., Blackshaw, Yang, J., Li, Y., Zuo, X., Zhen, Y., Yu, Y., Gao, L., 2008. Transient receptor potential
L.A., 2009. Role of TRPV1 in high-threshold rat colonic splanchnic afferents is ankyrin-1 participates in visceral hyperalgesia following experimental colitis.
revealed by inflammation. Neurosci. Lett. 459, 57–61. Neurosci. Lett. 440, 237–241.
Roberts, D.J., Hasler, W.L., Owyang, C., 1993. GABA mediation of the dual effects of Yoshimura, N., de Groat, W.C., 1997. Plasticity of Na+ channels in afferent neu-
somatostatin on guinea pig ileal myenteric cholinergic transmission. Am. J. Physiol. rones innervating rat urinary bladder following spinal cord injury. J. Physiol. 503
264, G953–G960. (Pt 2), 269–276.
Rong, W., Beyak, M.J., Bulmer, D.C.E., Jiang, W., Winchester, W., & Grundy, D., (5 A.D.) Yoshimura, N., de Groat, W.C., 1999. Increased excitability of afferent neurons innervating
Hydrogen sulphide activates small intestinal afferent fibres through a purinergic rat urinary bladder after chronic bladder inflammation. J. Neurosci. 19, 4644–4653.
mechanism. Gastroenterology 128[4 (2)], S1483. (Ref Type: Abstract). Yoshimura, N., White, G., Weight, F.F., de Groat, W.C., 1996. Different types of Na+ and
Rong, W., Spyer, K.M., Burnstock, G., 2002. Activation and sensitisation of low and high A-type K+ currents in dorsal root ganglion neurones innervating the rat urinary
threshold afferent fibres mediated by P2X receptors in the mouse urinary bladder. bladder. J. Physiol. 494 (Pt 1), 1–16.
J. Physiol. 541, 591–600. Zhu, J.X., Zhu, X.Y., Owyang, C., Li, Y., 2001. Intestinal serotonin acts as a paracrine
Rong, W., Hillsley, K., Davis, J.B., Hicks, G., Winchester, W.J., Grundy, D., 2004. Jejunal afferent substance to mediate vagal signal transmission evoked by luminal factors in the rat.
nerve sensitivity in wild-type and TRPV1 knockout mice. J. Physiol. 560, 867–881. J. Physiol. 530, 431–442.
Rong, W., Winchester, W.J., Grundy, D., 2007. Spontaneous hypersensitivity in mesen-
teric afferent nerves of mice deficient in the sst2 subtype of somatostatin receptor.
J. Physiol. 581, 779–786.

Вам также может понравиться