ArticlePDF AvailableLiterature Review

Hornby PJ. Central neurocircuitry associated with emesis. Am J Med111 (Suppl 8A):106S-112S

Authors:

Abstract and Figures

Ingestion of toxin, traumatic events, adverse drug reactions, and motion can all result in nausea and emesis. In addition, cyclic vomiting syndrome is quite prevalent in the pediatric population. Coordination of the various autonomic changes associated with emesis occurs at the level of the medulla oblongata of the hindbrain. Chemosensitive receptors detect emetic agents in the blood and relay this information by means of neurons in the area postrema to the adjacent nucleus tractus solitarius (NTS). Abdominal vagal afferents that detect intestinal luminal contents and gastric tone also terminate in the NTS (gelatinosus, commissural, and medial subnuclei). The NTS is viscerotopically organized into subnuclei that subserve diverse functions related to swallowing (subnucleus centralis), gastric sensation (subnucleus gelatinosus), laryngeal and pharyngeal sensation (intermediate and interstitial NTS), baroreceptor function (medial NTS), and respiration (ventrolateral NTS). Neurons from the NTS project to a central pattern generator (CPG), which coordinates the sequence of behaviors during emesis, as well as directly to diverse populations of neurons in the ventral medulla and hypothalamus. Thus, it is critical to realize that there is not an isolated "vomiting center," but rather groups of loosely organized neurons throughout the medulla that may be activated in sequence by a CPG. The newer antiemetic agents appear to block receptors in the peripheral endings of vagal afferents to reduce "perception" of emetic stimuli and/or act in the dorsal vagal complex. A primary site of action of 5-HT(3)-receptor antagonists is by means of the vagal afferents. Neurokinin-1 receptor (NK(1)R) antagonists are antiemetics, because they act at a site in the dorsal vagal complex. Part of their effectiveness may be the result of inhibition of the NK(1)R on vagal motor neurons to prevent fundic relaxation, which is a prodromal event essential for emesis. Delta(9)-tetrahydrocannabinol (Delta(9)-THC), the major psychoactive component of marijuana, can be therapeutically useful as an antiemetic. The site of action of Delta(9)-THC is on cannabinoid CB1 receptors in the dorsal vagal complex. However, it decreases fundic tone and antral motility. It is not easy to predict the potential antiemetic effects of drugs that alter motility. Although antiemetic drugs are available for management of acute chemotherapeutic-induced emesis, few treatments are effective for delayed emesis or cyclic vomiting syndrome.
Approximate location of neurons in hindbrain medulla controlling behavior during emesis. Only neuronal structures and regions in which there is increased c-fos activation after an emetic stimulus in decerebrate cats are labeled. To schematically represent the data, locations of labeled regions may not be anatomically precise. Lines indicate primary function of areas activated; for simplicity, neural connections involved are not shown. (Inset) enlargement of the dorsal vagal complex (DVC) illustrating that abdominal vagal afferents terminate in the gelatinosus and medial nucleus tractus solitarius (NTS) subnuclei as well as in the more caudal commisural NTS (lower right). Information from vagal afferents and the area postrema is transferred to the NTS, where the subnuclei illustrated subserve diverse functions (as described elsewhere in this supplement 4,5 ). This information is also relayed to neurons in the central pattern generator (CPG) and surrounding regions. Neurons controlling respiratory behavior are located in the Bötzinger/ventral respiratory group (Böt/VRG). Premotor neurons controlling the larynx and pharynx are located in the nearby rostral nucleus ambiguus/retrofacial nucleus (nAmb/RFN). Changes in neurohypophyseal hormone release from the hypothalamus are relayed from catecholaminergic groups in the ventrolateral medulla (R/CVLM) and from the NTS. Blood pressure and heart rate are controlled by the R/CVLM and nAmb, respectively. Extensive connections between the NTS and dorsal motor nucleus (DMN) of the vagus control motor function of the lower esophageal sphincter (LES) and stomach. AP area postrema; cen subnucleus centralis; com commissural subnucleus of NTS; gel subnucleus gelatinosus; int intermediate subnucleus; is interstitial subnucleus; med medial NTS; mlf medial longitudinal fasciculus; OXY/AVP oxytocin/arginine vasopressin; R/CVLM rostral/caudal ventrolateral medulla; RM raphe magus; TS tractus solitarius; V4 fourth ventricle; vl ventrolateral subnucleus of NTS; X11 hypoglassal nucleus. (Modified with permission from J Neurosci. 3 )
… 
Content may be subject to copyright.
Central Neurocircuitry Associated with Emesis
Pamela J. Hornby, PhD
Ingestion of toxin, traumatic events, adverse drug
reactions, and motion can all result in nausea and
emesis. In addition, cyclic vomiting syndrome is
quite prevalent in the pediatric population. Coordina-
tion of the various autonomic changes associated
with emesis occurs at the level of the medulla oblon-
gata of the hindbrain. Chemosensitive receptors de-
tect emetic agents in the blood and relay this infor-
mation by means of neurons in the area postrema to
the adjacent nucleus tractus solitarius (NTS). Ab-
dominal vagal afferents that detect intestinal luminal
contents and gastric tone also terminate in the NTS
(gelatinosus, commissural, and medial subnuclei).
The NTS is viscerotopically organized into subnuclei
that subserve diverse functions related to swallow-
ing (subnucleus centralis), gastric sensation (subnu-
cleus gelatinosus), laryngeal and pharyngeal sensa-
tion (intermediate and interstitial NTS), baroreceptor
function (medial NTS), and respiration (ventrolateral
NTS). Neurons from the NTS project to a central
pattern generator (CPG), which coordinates the se-
quence of behaviors during emesis, as well as di-
rectly to diverse populations of neurons in the ventral
medulla and hypothalamus. Thus, it is critical to re-
alize that there is not an isolated “vomiting center,”
but rather groups of loosely organized neurons
throughout the medulla that may be activated in se-
quence by a CPG.
The newer antiemetic agents appear to block re-
ceptors in the peripheral endings of vagal afferents
to reduce “perception” of emetic stimuli and/or act in
the dorsal vagal complex. A primary site of action of
5-HT
3
-receptor antagonists is by means of the vagal
afferents. Neurokinin-1 receptor (NK
1
R) antagonists
are antiemetics, because they act at a site in the
dorsal vagal complex. Part of their effectiveness may
be the result of inhibition of the NK
1
R on vagal motor
neurons to prevent fundic relaxation, which is a pro-
dromal event essential for emesis. Delta
9
-tetrahy-
drocannabinol (
9
-THC), the major psychoactive
component of marijuana, can be therapeutically use-
ful as an antiemetic. The site of action of
9
-THC is
on cannabinoid CB1 receptors in the dorsal vagal
complex. However, it decreases fundic tone and an-
tral motility. It is not easy to predict the potential
antiemetic effects of drugs that alter motility. Al-
though antiemetic drugs are available for manage-
ment of acute chemotherapeutic-induced emesis,
few treatments are effective for delayed emesis or
cyclic vomiting syndrome. Am J Med. 2001;111(8A):
106S–112S. © 2001 by Excerpta Medica, Inc.
Ingestion of toxin, “cephalic” responses to traumatic
or repugnant events, and motion can result in nausea
and emesis. For many patients, treatment with opioid
analgesics, anticancer agents, and other drugs also may
cause nausea and emesis. In addition, cyclic vomiting
syndrome (CVS) is present in approximately 2% of the
pediatric population.
1
CVS is characterized by episodes
of frequent and intractable vomiting followed by symp-
tom-free periods that vary in duration.
Nausea cannot be studied in nonhumans, although be-
haviors such as excessive salivation and swallowing pro-
vide insight into the neural control of this unpleasant
sensation. On the other hand, vomiting that comprises
retching and expulsion phases can easily be studied in
experimental models. Because emesis occurs infre-
quently, toxins such as apomorphine, anticancer agents,
or irradiation can be used to study neural control of these
complex events. During the retching phase, muscles of
the diaphragm and abdomen simultaneously contract or
relax. During expulsion, there is prolonged contraction
of the abdominal muscles that is coordinated with the
activity of the intercostal muscles and the muscles of the
larynx and pharynx. The glottis is closed and the soft pal-
ate elevated. In the intestines, a retrograde giant contrac-
tion occurs and the gastric fundus relaxes, which is an
essential prodromal event for emesis.
2
Pulse rate and
breathing increase and sweating occurs.
HINDBRAIN NEUROCIRCUITRY
ASSOCIATED WITH EMESIS
Coordination of this extraordinary combination of auto-
nomic changes occurs at the level of the medulla oblon-
gata of the hindbrain (Figure 1). The region that is essen-
tial for coordinating these behaviors is located between
the level of the obex (opening of central canal into fourth
ventricle) to the level of the rostral portion (compact
zone) of the nucleus ambiguus, which is termed the ret-
rofacial nucleus in cats (Figure 2).
3–5
Thus, output neu-
rons that control the muscles involved in emesis are scat-
tered throughout the medulla oblongata. What is distinc-
tive about vomiting is that these neurons must be
From the Department of Pharmacology and Neuroscience Center of
Excellence, Louisiana State University Health Sciences Center, New Or-
leans, Louisiana, USA.
This work was supported by the National Institute of Diabetes and
Digestive and Kidney Diseases (Grant No. PHS 42714).
Requests for reprints should be addressed to Pamela J. Hornby, PhD,
Department of Pharmacology, Louisiana State University Health Sci-
ences Center, 1901 Perdido Street, New Orleans, Louisiana 70112.
106S © 2001 by Excerpta Medica, Inc. 0002-9343/01/$20.00
All rights reserved. PII S0002-9343(01)00849-X
activated in the appropriate sequence. This sequence of
events can best be understood in terms of a pattern gen-
erator,rather than a vomiting centerper se.
It is beyond the scope of this brief overview to describe
all of the neurons that coordinate control of the dia-
phragm, inspiration, blood pressure, heart rate, larynx,
pharynx, tongue, lower esophageal sphincter (LES), and
gastric fundus. In addition, a considerable body of litera-
ture already describes the neural circuitry underlying re-
spiratory and abdominal pressure changes associated
with emesis.
6
Therefore, this article will focus on neural
control of upper gastrointestinal changes and possible
sites of action of antiemetic drugs. It should become ap-
parent that considerable overlap exists in the neurons
that control emesis and those involved in other supra-
esophageal functions, such as swallowing. Gaps in our
knowledge include, for example, information on how the
brain and enteric nervous system coordinate the giant
retrograde contraction that extends through the intes-
tines to the level of the gastric corpus.
2,7
Wang and Borison
8
first proposed the idea of a vomit-
ing center. However, some of their original observations
have not been supported by more recent studies. For ex-
ample, they showed that vomiting could be induced by
stimulation of the dorsolateral medulla in cats,
8
but other
investigators have been unable to find a discrete site from
which they could consistently elicit vomiting.
9
Moreover,
vomiting could still be induced after neuronal cell bodies
in the dorsomedial medulla were selectively lesioned.
10
Thus, the simple concept of a vomiting center that could
be readily manipulated pharmacologically or surgically
has not been upheld.
What has been upheld by subsequent research, how-
ever, is the original observation that the integrity of the
abdominal vagus is essential for emesis.
11
In ferrets, stim-
ulation of mucosal chemoreceptors in the stomach or
duodenum by luminal hydrochloride or hypertonic sa-
line results in long latency and sudden increases in vagal
efferent discharge associated with the prodrome of vom-
iting.
12
Thus, signals associated with luminal contents are
detected by vagal afferent chemoreceptors in the mucosa
and relayed to the hindbrain by a rapid and distinctive
firing pattern from vagal motor nerve fibers. Interest-
ingly, successful treatment of intractable epilepsy in pa-
tients has been accomplished by repeated stimulation of
the left vagus nerve through implanted electrodes, but
nausea and vomiting do not occur with this treatment.
13
Thus, vagal activation alone is not sufficient to trigger
emesis.
The other idea proposed by earlier studies that has
passed the test of time is that a major site involved in
triggering emesis is near the area postrema. Chemorecep-
tors in the area postrema, which are outside of the blood–
brain barrier, are sensitive to circulating emetic agents,
such as apomorphine or cytotoxic drugs.
14
An example of
how these peripheral chemoreceptors are involved in
triggering emesis comes from observation of patients
with Parkinson’s disease who receive L-dopa. Emesis
caused by dopamine receptor stimulation is less likely
when L-dopa is coadministered with a peripherally acting
dopa decarboxylase inhibitor. Thus, peripherally active
dopamine, converted from L-dopa, triggers emesis. In
contrast to drug-induced emesis, the area postrema is less
Figure 1. Some triggering factors in emesis that impinge on the emetic circuitry in the hindbrain medulla from the level of the
medullary–spinal transition to the pons.
A Symposium: Central Nervous System Emetic Circuitry/Hornby
December 3, 2001 THE AMERICAN JOURNAL OF MEDICINEVolume 111 (8A) 107S
involved in motion sickness,
15
which is caused by stimu-
lation of labyrinthine end organs.
Some of the hindbrain neurocircuitry of neuronal
populations controlling behaviors related to emesis are
shown in Figure 2. Activation of gastric vagal afferents
stimulates neurons in the area postrema
16
and in the
nearby nucleus tractus solitarius (NTS), specically in the
subnucleus gelatinosus neurons.
17
In this subnucleus of
the NTS, there is a marked concentration of gastric affer-
ent input. Emetic agents activate a similar distribution of
neurons in the NTS of cats
3
and ferrets.
18
Different stud-
ies have either reported the presence or absence of c-fos
activation in the subnucleus gelatinosus after emetic or
abdominal stimuli, although this partly may be caused by
the paucity of neurons in this subnucleus, which is pre-
dominantly composed of axodendritic synapses related
to gastric reexes.
19
Neurons activated in the dorsal vagal
complex (DVC), in addition to the subnucleus gelatino-
sus, are located in areas controlling swallowing (subnu-
cleus centralis of the NTS), baroreceptor reexes (medial
NTS), respiration (ventrolateral NTS), and tone/motility
of the stomach and LES (dorsal motor nucleus of the
vagus). Recently, we have shown that approximately half
of the preganglionic motor neurons in the dorsal motor
nucleus of the vagus that innervate the LES also innervate
the gastric fundus.
20
This may have relevance for emesis,
as both fundic relaxation and LES relaxation precede
emesis.
7
Widespread areas of activated cells are also noted in the
ventral medulla after emetic stimuli (Figure 2). These re-
gions include premotor neurons in the compact zone of
the nucleus ambiguus (retrofacial nucleus) that innervate
the larynx and pharynx; para-ambigual expiratory-re-
lated neurons that control respiration; ventrolateral med-
ullary neurons that control sympathetic outow to main-
tain blood pressure; and parasympathetic neurons in the
nucleus ambiguus that innervate the heart. Projections
from the NTS to the ventrolateral medulla may be partic-
Figure 2. Approximate location of neurons in hindbrain medulla controlling behavior during emesis. Only neuronal structures
and regions in which there is increased c-fos activation after an emetic stimulus in decerebrate cats are labeled. To schematically
represent the data, locations of labeled regions may not be anatomically precise. Lines indicate primary function of areas activated;
for simplicity, neural connections involved are not shown. (Inset) enlargement of the dorsal vagal complex (DVC) illustrating that
abdominal vagal afferents terminate in the gelatinosus and medial nucleus tractus solitarius (NTS) subnuclei as well as in the more
caudal commisural NTS (lower right). Information from vagal afferents and the area postrema is transferred to the NTS, where the
subnuclei illustrated subserve diverse functions (as described elsewhere in this supplement
4,5
). This information is also relayed to
neurons in the central pattern generator (CPG) and surrounding regions. Neurons controlling respiratory behavior are located in the
Bo¨tzinger/ventral respiratory group (Bo¨t/VRG). Premotor neurons controlling the larynx and pharynx are located in the nearby
rostral nucleus ambiguus/retrofacial nucleus (nAmb/RFN). Changes in neurohypophyseal hormone release from the hypothalamus
are relayed from catecholaminergic groups in the ventrolateral medulla (R/CVLM) and from the NTS. Blood pressure and heart rate
are controlled by the R/CVLM and nAmb, respectively. Extensive connections between the NTS and dorsal motor nucleus (DMN)
of the vagus control motor function of the lower esophageal sphincter (LES) and stomach. AP area postrema; cen subnucleus
centralis; com commissural subnucleus of NTS; gel subnucleus gelatinosus; int intermediate subnucleus; is interstitial
subnucleus; med medial NTS; mlf medial longitudinal fasciculus; OXY/AVP oxytocin/arginine vasopressin; R/CVLM
rostral/caudal ventrolateral medulla; RM raphe magus; TS tractus solitarius; V4 fourth ventricle; vl ventrolateral subnu-
cleus of NTS; X11 hypoglassal nucleus. (Modied with permission from J Neurosci.
3
)
A Symposium: Central Nervous System Emetic Circuitry/Hornby
108S December 3, 2001 THE AMERICAN JOURNAL OF MEDICINEVolume 111 (8A)
ularly important for mediating the respiratory motor
components of vomiting.
21,22
Neurons in the rostral por-
tion of the nucleus ambiguus (at the level just caudal to
the facial nucleus, hence termed retrofacial nucleus in
cats) have ring characteristics consistent with a possible
role as central pattern generators.
23,24
Vasopressin and oxytocin levels increase in humans
experiencing nausea from both illusory self-motion and a
number of emetic-producing treatments. Dopamine an-
tagonists abolish both nausea and the increase in vaso-
pressin.
25
A vasopressin V1 antagonist has demonstrated
complete blockade of emesis and other signicant symp-
toms in squirrel monkeys.
15
In contrast to vasopressin,
oxytocin release in the hypothalamus can be triggered by
normal eating and can signal sensations of both satiety
and malaise behavior in rats.
26
Discharge ring of oxyto-
cin neurons, but not vasopressin neurons, accompanies
gastric distention and cholecystokinin administration.
27
The pathway by which this occurs can be directly from
the NTS to the magnocellular hypothalamic neurons, and
by means of catecholaminergic neurons in the ventrolat-
eral medulla (Figure 2). These are identied as part of the
central pathway by which afferent abdominal vagal stim-
ulation increases plasma vasopressin and also increases
arterial pressure.
28
Overall, as Verbalis et al
26
have con-
cluded, increasing levels of neurohypophyseal hormones
are a marker of a disinclination to eat, with higher levels
indicating malaise rather than satiety.
Expulsion has relatively little to do with gastrointesti-
nal function; rather, it is a response to changes in intra-
abdominal and intrathoracic pressure generated by the
respiratory muscles. The respiratory-related components
of ctive vomiting can be abolished by lesions of the lat-
eral medulla at the level of the retrofacial nucleus, where
respiratory premotor and motor neurons exist.
10
Phrenic
motor neurons controlling the diaphragm are largely
conned to the C5C7 levels of spinal cord location. In-
puts to these neurons during inspiratory behaviors
unique to emesis arise not only from premotor neurons
in the ventral medulla nucleus ambiguus region, but also
from neurons in the midline of the medulla.
29
SITE OF ACTION OF NEWER
ANTIEMETIC DRUGS
It is becoming clear that the vagal afferents are a major
site of action of antiemetic drugs (Figure 3). The anti-
emetic action of the 5-HT
3
-receptor antagonists, such as
granisetron and odansetron, are primarily the result of
Figure 3. Selected drugs that affect emesis and their site(s) of action (if known).
2
Radrenergic
2
-receptor; CPG central
pattern generator; D
2
Rdopamine
2
-receptor;
9
-THC ⫽⌬
9
-tetrahydrocannabinol; DVC dorsal vagal complex; 5-HT
serotonin; H histamine; mR cholinergic muscarinic receptor; NK neurokinin; R receptor.
A Symposium: Central Nervous System Emetic Circuitry/Hornby
December 3, 2001 THE AMERICAN JOURNAL OF MEDICINEVolume 111 (8A) 109S
their effects on the abdominal vagal afferents.
30
This is
because cytotoxic agents increase the release of 5-HT
from enterochromafn cells in the small intestinal mu-
cosa, and 5-HT subsequently activates 5-HT
3
receptors
on vagal abdominal afferents.
31,32
The discharge of affer-
ent vagal bers to 5-HT is completely abolished by
5-HT
3
-receptor antagonists.
33
However, these agents are
less effective in delayed vomiting. Hasler
34
also has re-
viewed data suggesting that 5-HT
4
agonists may amelio-
rate nausea in patients with gastroparesis and functional
dyspepsia because of prokinetic effects that stimulate gas-
tric emptying.
It is assumed that some underlying causes of vomiting
result from impaired gastrointestinal motor activity.
Disruption of gastric slow-wave dysrhythmias (resulting
in both tachygastria and bradygastria) has been associ-
ated with nausea and emesis related to motion sick-
ness
35
and pregnancy hyperemesis.
36
Neurokinin-1 re-
ceptor (NK
1
R) antagonists have been shown to prevent
emesis in many species, including humans,
37
and show
promise in treating delayed chemotherapy-evoked eme-
sis. Furthermore, animal studies have indicated a broad
spectrum of action for NK
1
R antagonists in treating di-
verse causes of nausea and vomiting.
38
The antiemetic
properties of NK
1
R antagonists are thought to act in the
DVC,
3941
but the specic site of action is not known.
They may act in the area postrema, because HSP-117 and
CP-99,994 (NK
1
R antagonists) applied to the area pos-
trema have been shown to decrease expulsion and retch-
ing episodes induced by morphine and copper sulfate.
42
However, in these particular studies, diffusion of the
drugs may have affected other regions of the DVC. It is
unlikely that NK
1
R antagonists prevent emesis at the level
of the primary afferent inputs to the NTS because intra-
venous GR-205171 does not affect the response of medial
NTS neurons to vagal stimulation even though retching
was abolished.
43
We have recently shown that the NK
1
Ris
highly expressed in vagal motor neurons
44
and NK
1
Rac
-
tivation in this region potently evokes gastric fundic re-
laxation.
45
Because fundic relaxation is a prodromal
event essential for emesis, it is attractive to speculate that
these antagonists inhibit fundic relaxation by blocking
the NK
1
R on preganglionic neurons in the dorsal motor
nucleus of the vagus. In summary, the antiemetic site of
action of NK
1
R antagonists in the DVC is elusive but may
involve vagal motor neurons that control fundic relax-
ation.
The major psychoactive component of marijuana,
9
-
tetrahydrocannabinol (
9
-THC), inhibits gastrointesti-
nal transit.
46
It is also used as an antiemetic
47
and syn-
thetic cannabinoids dronabinol and nabilone have been
produced for this use. Delta
9
-THC inhibits gastric emp-
tying in humans,
48
and we found that in rats
9
-THC
administered peripherally decreased fundic tone and an-
tral motility by means of cannabinoid CB1 receptors.
49
This effect is completely abolished by vagotomy and,
when the drug is applied to the surface of the medulla
above the DVC, at a dose that is ineffective when given
intravenously, it decreases fundic tone and antral motil-
ity.
49
Cannabinoid receptor mRNA is located in the DVC
of the rat,
50
and therefore
9
-THC may act partly in the
DVC to decrease fundic tone. This may be helpful to al-
low the stomach to accommodate food intake, which is
a problem for patients with functional dyspepsia, but it is
an enigma how
9
-THCevoked gastrointestinal stasis
and fundic relaxation may contribute to its antiemetic
effects.
Cyclic Vomiting Syndrome
The cause of CVS is incompletely understood. Its onset is
generally in children younger than 5 years of age and
results in approximately 12 attacks per year. In some
cases, it ceases with adolescence.
1
CVS may be related to
migraine headaches. Some success has been noted with
use of the antimigraine drug sumatriptan (a 5-HT
1D
ag-
onist) to reduce the severity of cyclic vomiting attacks.
38
Paradoxically, sumatriptan is not as effective for migraine
in children as it is in adults.
51,52
Vomiting may be stress
initiated, and patients try to avoid the trigger for their
attacks.
CONCLUSIONS
The locations of neurons that coordinate the bodily func-
tions associated with emesis are spread throughout the
medulla, supporting the notion that a central pattern
generator coordinates the sequence of behaviors during
emesis. This region receives indirect input from both the
area postrema and abdominal vagus by means of the
NTS. Despite this information, there is little evidence that
antiemetic agents act directly on the central pattern gen-
erator. Neither is there any indication that drugs that alter
inspiratory behaviors associated with emesis would be
effective antiemetics. Overall, the newer antiemetic
agents act on pathways related to vagal gastric function
either at the peripheral ending of abdominal vagal affer-
ents or in the area postrema and DVC. Thus, 5-HT
3
receptor antagonists are an effective and rational ap-
proach for preventing vagal afferent signaling to the
hindbrain. It is unclear whether alteration of fundic re-
laxation is a component of the antiemetic effects of such
agents as dronabinol or NK
1
R antagonists, although cer-
tainly these agents act in the DVC to mediate their effects.
Possibly, drugs that alter motility may be effective anti-
nausea/antiemetic agents. An enigmatic facet of this ap-
proach is the fact that both tachygastria and bradygastria
are associated with nausea. Therefore, it is not easy to
predict the effects of drugs that alter motility on nausea
and vomiting. For example, a motilin receptor agonist,
ABT-229, with prokinetic effects by means of stimulation
of enteric nerves or on smooth muscle
53
worsens the
A Symposium: Central Nervous System Emetic Circuitry/Hornby
110S December 3, 2001 THE AMERICAN JOURNAL OF MEDICINEVolume 111 (8A)
symptoms of nausea and vomiting in patients with func-
tional dyspepsia and seems to be of limited usefulness.
54
Finally, although antiemetic drugs are available that are
effective for acute chemotherapeutic-induced emesis,
there are few effective treatments for delayed emesis or
CVS.
ACKNOWLEDGMENT
I appreciate the editorial assistance of Melissa Burmeister.
REFERENCES
1. Forbes D. Cyclic vomiting syndrome. In: Hyman PE, ed.
Pediatric Functional Gastrointestinal Disorders. New York:
Academy Professional Information Services, 1999:5.1
5.12.
2. Lang IM. Digestive tract motor correlates of vomiting and
nausea. Can J Physiol Pharmacol. 1990;68:242253.
3. Miller AD, Ruggiero DA. Emetic reflex arc revealed by ex-
pression of the immediate-early gene c-fos in the cat.
J Neurosci. 1994;14:871–888.
4. Altschuler SM. Laryngeal and respiratory protective re-
flexes. Am J Med. 2001;111(suppl):90S–94S.
5. Bieger D. Rhombencephalic pathways and neurotransmit-
ters controlling deglutition. Am J Med. 2001;111(suppl):
85S–89S.
6. Miller AD. Central mechanisms of vomiting. Dig Dis Sci.
1999;44:39S–43S.
7. Lang IM, Sarna SK, Dodds WJ. Pharyngeal, esophageal,
and proximal gastric responses associated with vomiting.
Am J Physiol. 1993;265:G963–G972.
8. Wang SC, Borison HL. The vomiting center: a critical ex-
perimental analysis. Arch Neurol Psychiatry. 1950;63:928
941.
9. Miller AD, Wilson VJ. “Vomiting center” reanalyzed: an elec-
trical stimulation study. Brain Res. 1983;270:154–158.
10. Miller AD, Nonaka S, Jakus J. Brain areas essential or
non-essential for emesis. Brain Res. 1994;647:255–264.
11. Miller FR. On gastric sensation. J Physiol. 1910;41:409
415.
12. Blackshaw LA, Grundy D, Scratcherd T. Involvement of
gastrointestinal mechano- and intestinal chemoreceptors in
vagal reflexes: an electrophysiological study. J Auton Nerv
Syst. 1987;18:225–234.
13. Binnie CD. Vagus nerve stimulation for epilepsy: a review.
Seizure. 2000;9:161–169.
14. Borison HL. Area postrema: chemoreceptor circumven-
tricular organ of the medulla oblongata. Prog Neurobiol.
1989;32:351–390.
15. Kohl RL, MacDonald S. New pharmacologic approaches to
the prevention of space/motion sickness. J Clin Pharmacol.
1991;31:934–946.
16. Yuan CS, Barber WD. Area postrema: gastric vagal input
from proximal stomach and interactions with nucleus trac-
tus solitarius in cat. Brain Res Bull. 1993;30:119–125.
17. Boissonade FM, Sharkey KA, Davison JS. Fos expression
in ferret dorsal vagal complex after peripheral emetic stim-
uli. Am J Physiol. 1994;266:R1118–R1126.
18. Reynolds DJ, Barber NA, Grahame-Smith DG, Leslie RA.
Cisplatin-evoked induction of c-fos protein in the brainstem
of the ferret: the effect of cervical vagotomy and the anti-
emetic 5-HT
3
receptor antagonist granisetron (BRL 43694).
Brain Res. 1991;565:231–236.
19. Rinaman L, Card JP, Schwaber JS, Miselis RR. Ultrastruc-
tural demonstration of a gastric monosynaptic vagal circuit
in the nucleus of the solitary tract in rat. J Neurosci. 1989;
9:1985–1996.
20. Hyland NP, Abrahams TP, Fuchs K, et al. Organization and
neurochemistry of vagal preganglionic neurons innervating
the lower esophageal sphincter in ferrets. J Comp Neurol.
2001;430:222–234.
21. Fukuda H, Koga T. The Botzinger complex as the pattern
generator for retching and vomiting in the dog. Neurosci
Res. 1991;12:471–485.
22. Koga T, Qu R, Fukuda H. The central pattern generator for
vomiting may exist in the reticular area dorsomedial to the
retrofacial nucleus in dogs. Exp Brain Res. 1998;118:139
147.
23. Fukuda H, Koga T. Non-respiratory neurons in the Botz-
inger complex exhibiting appropriate firing patterns to gen-
erate the emetic act in dogs. Neurosci Res. 1992;14:180
194.
24. Koga T, Fukuda H. Neurons in the nucleus of the solitary
tract mediating inputs from emetic vagal afferents and the
area postrema to the pattern generator for the emetic act in
dogs. Neurosci Res. 1992;14:166–179.
25. Rowe JW, Shelton RL, Helderman JH, et al. Influence of the
emetic reflex on vasopressin release in man. Kidney Int.
1979;16:729–735.
26. Verbalis JG, McCann MJ, McHale CM, Stricker EM. Oxy-
tocin secretion in response to cholecystokinin and food:
differentiation of nausea from satiety. Science. 1986;232:
1417–1419.
27. Renaud LP, Tang M, McCann MJ, et al. Cholecystokinin
and gastric distension activate oxytocinergic cells in rat
hypothalamus. Am J Physiol. 1987;253:R661–R665.
28. Gieroba ZJ, Blessing WW. Fos-containing neurons in me-
dulla and pons after unilateral stimulation of the afferent
abdominal vagus in conscious rabbits. Neuroscience.
1994;59:851–858.
29. Yates BJ, Smail JA, Stocker SD, Card JP. Transneuronal
tracing of neural pathways controlling activity of diaphragm
motoneurons in the ferret. Neuroscience. 1999;90:1501–
1513.
30. Andrews PL, Davis CJ, Bingham S, et al. The abdominal
visceral innervation and the emetic reflex: pathways, phar-
macology, and plasticity. Can J Physiol Pharmacol. 1990;
68:325–345.
31. Hillsley K, Grundy D. Sensitivity to 5-hydroxytryptamine in
different afferent subpopulations within mesenteric nerves
supplying the rat jejunum. J Physiol (Lond). 1998;509:717–
727.
32. Hillsley K, Kirkup AJ, Grundy D. Direct and indirect actions
of 5-hydroxytryptamine on the discharge of mesenteric
afferent fibres innervating the rat jejunum. J Physiol (Lond).
1998;506:551–561.
33. Blackshaw LA, Grundy D. Effects of 5-hydroxytryptamine
on discharge of vagal mucosal afferent fibres from the
upper gastrointestinal tract of the ferret. J Auton Nerv Syst.
1993;45:41–50.
34. Hasler WL. Serotonin receptor physiology: relation to eme-
sis. Dig Dis Sci. 1999;44(suppl):108S–113S.
35. Kim MS, Chey WD, Owyang C, Hasler WL. Role of plasma
vasopressin as a mediator of nausea and gastric slow wave
dysrhythmias in motion sickness. Am J Physiol. 1997;272:
G853–G862.
36. Jednak MA, Shadigian EM, Kim MS, et al. Protein meals
reduce nausea and gastric slow wave dysrhythmic activity
in first trimester pregnancy. Am J Physiol. 1999;277:G855–
G861.
A Symposium: Central Nervous System Emetic Circuitry/Hornby
December 3, 2001 THE AMERICAN JOURNAL OF MEDICINEVolume 111 (8A) 111S
37. Navari RM, Reinhardt RR, Gralla RJ, et al. Reduction of
cisplatin-induced emesis by a selective neurokinin-1-re-
ceptor antagonist. L-754,030 Antiemetic Trials Group.
N Engl J Med. 1999;340:190195.
38. Ladabaum U, Hasler WL. Novel approaches to the treat-
ment of nausea and vomiting. Dig Dis. 1999;17:125132.
39. Rudd JA, Ngan MP, Wai MK. Inhibition of emesis by tachy-
kinin NK1 receptor antagonists in Suncus murinus (house
musk shrew). Eur J Pharmacol. 1999;366:243252.
40. Tattersall FD, Rycroft W, Francis B, et al. Tachykinin NK1
receptor antagonists act centrally to inhibit emesis induced
by the chemotherapeutic agent cisplatin in ferrets. Neuro-
pharmacology. 1996;35:11211129.
41. Watson JW, Gonsalves SF, Fossa AA, et al. The antiemetic
effects of CP-99,994 in the ferret and the dog: role of the
NK1 receptor. Br J Pharmacol. 1995;115:8494.
42. Ariumi H, Saito R, Nago S, et al. The role of tachykinin NK-1
receptors in the area postrema of ferrets in emesis. Neuro-
sci Lett. 2000;286:123126.
43. Fukuda H, Koga T, Furukawa N, et al. The tachykinin NK1
receptor antagonist GR205171 prevents vagal stimulation-
induced retching but not neuronal transmission from
emetic vagal afferents to solitary nucleus neurons in dogs.
Brain Res. 1998;802:221231.
44. Dixon MK, Nathan NA, Hornby PJ. Immunocytochemical
distribution of neurokinin 1 receptor in rat dorsal vagal
complex. Peptides. 1998;19:913923.
45. Krowicki ZK, Hornby PJ. Substance P in the dorsal motor
nucleus of the vagus evokes gastric motor inhibition via
neurokinin 1 receptor in rat. J Pharmacol Exp Ther. 2000;
293:214221.
46. Shook JE, Burks TF. Psychoactive cannabinoids reduce
gastrointestinal propulsion and motility in rodents. J Phar-
macol Exp Ther. 1989;249:444449.
47. Gonzalez-Rosales F, Walsh D. Intractable nausea and vom-
iting due to gastrointestinal mucosal metastases relieved
by tetrahydrocannabinol (dronabinol). J Pain Symptom
Manage. 1997;14:311314.
48. McCallum RW, Soykan I, Sridhar KR, et al. Delta-9-tetra-
hydrocannabinol delays the gastric emptying of solid food
in humans: a double-blind, randomized study. Aliment
Pharmacol Ther. 1999;13:7780.
49. Krowicki ZK, Moerschbaecher JM, Winsauer PJ, et al. Del-
ta-9-tetrahydrocannabinol inhibits gastric motility in the rat
through cannabinoid CB1 receptors. Eur J Pharmacol.
1999;371:187196.
50. Matsuda LA, Bonner TI, Lolait SJ. Localization of cannabi-
noid receptor mRNA in rat brain. J Comp Neurol. 1993;327:
535550.
51. Hamalainen ML, Hoppu K, Santavuori P. Sumatriptan for
migraine attacks in children: a randomized placebo-con-
trolled study. Do children with migraine respond to oral
sumatriptan differently from adults? Neurology. 1997;48:
11001103.
52. Rederich G, Rapoport A, Cutler N, et al. Oral sumatriptan
for the long-term treatment of migraine: clinical ndings.
Neurology. 1995;45:S15S20.
53. Sarna SK, Gonzalez A, Ryan RP. Enteric locus of action of
prokinetics: ABT-229, motilin, and erythromycin. Am J
Physiol. 2000;278:G744G752.
54. Van Herwaarden MA, Samsom M, Van Nispen CH, et al.
The effect of motilin agonist ABT-229 on gastro-oesopha-
geal reux, oesophageal motility and lower oesophageal
sphincter characteristics in GERD patients. Aliment Phar-
macol Ther. 2000;14:453462.
A Symposium: Central Nervous System Emetic Circuitry/Hornby
112S December 3, 2001 THE AMERICAN JOURNAL OF MEDICINEVolume 111 (8A)
Article
Migraine is a form of primary headache that affects at least 10% of the world’s population. In addition to recommendations for modifying the patient’s lifestyle, migraine management involves stopping an attack that’s already occurred and/or preventing its occurrence. In the abortive treatment of this cephalalgia, both non-specific (eg, non-opioid analgesics) and specific pharmacological agents, can be used. The latter include, in particular, serotonergic drugs of the classes of triptans (selective 5-HT1B/1D receptor agonists), ditans (selective 5-HT1F-mimetics), and ergot alkaloids (non-selective modulators of various 5-HT receptor subtypes). The review discusses the currently availably results of numerous basic and applied studies of these drug groups, in which the neuronal and vascular components of their antimigraine pharmacodynamics were identified. A significant part of the information was obtained in vivo on the various experimental models of migraine based on the trigeminovascular theory of its pathogenesis. Other data are the results of ex vivo studies on isolated tissues and cell cultures. When analyzing these experimental results, evidence is provided in favor of similar mechanisms for realizing the antimigraine potential of all representatives belonging to the pharmacological classes listed, the neurotropic activity of which prevails over their direct intervention in vascular tone. At the same time, special attention is paid to the controversial and debatable issues in this area, the successful solution of which is a key to further progress in the pharmacotherapy of migraine.
Article
Full-text available
Background/Aims We examined the involvement of cholecystokinin (CCK) in the exacerbation of indomethacin (IND)-induced gastric antral ulcers by gastroparesis caused by atropine or dopamine in mice. Methods Male mice were fed for 2 h (re-feeding) following a 22-h fast. Indomethacin (IND; 10 mg/kg, s.c.) was administered after re-feeding; gastric lesions were examined 24 h after IND treatment. In another experiment, mice were fed for 2 h after a 22-h fast, after which the stomachs were removed 1.5 h after the end of the feeding period. Antral lesions, the amount of gastric contents, and the gastric luminal bile acids concentration were measured with or without the administration of the pro- and antimotility drugs CCK-octapeptide (CCK-8), atropine, dopamine, SR57227 (5-HT3 receptor agonist), apomorphine, lorglumide (CCK1 receptor antagonist), ondansetron, and haloperidol alone and in combination. Results IND produced severe lesions only in the gastric antrum in re-fed mice. CCK-8, atropine, dopamine, SR57227 and apomorphine administered just after re-feeding increased bile reflux and worsened IND-induced antral lesions. These effects were significantly prevented by pretreatment with lorglumide. Although atropine and dopamine also increased the amount of gastric content, lorglumide had no effect on the delayed gastric emptying provoked by atropine and dopamine. Both ondansetron and haloperidol significantly inhibited the increase of bile reflux and the exacerbation of antral lesions induced by atropine and dopamine, respectively, but did not affect the effects of CCK-8. Conclusions These results suggest that CCK-CCK1 receptor signal increases bile reflux during gastroparesis induced by atropine and dopamine, exacerbating IND-induced antral ulcers. Graphical Abstract
Article
Full-text available
The emergency department (ED) is at times the only place where patients can turn for symptom relief. Patients of all ages may turn to the ED for help with the management of end-of-life (EOL) and palliative care (PC) symptoms. Emergency medicine (EM) is a specialty that manages disease-directed treatment for a variety of acute conditions. In contrast, EOL and PC are focused on improving quality of life. Patients with serious illness, even hospice patients, present to the ED in increasing numbers for symptom management. It has become essential for emergency physicians to care for patients who are not seeking life-sustaining measures but instead need quality-of-life interventions. The development of a clear, concise review of the most common acute symptoms can provide a framework for EM physicians to adequately address the needs of patients at the EOL. Here, we discuss three cases that highlight the management of five of the most common EOL and PC presentations to the ED.
Article
Increasing the therapeutic potential and reducing the side effects of U.S. Food and Drug Administration–approved glucagon-like peptide-1 receptor (GLP-1R) agonists used to treat obesity require complete characterization of the central mechanisms that mediate both the food intake-suppressive and illness-like effects of GLP-1R signaling. Our studies, in the rat, demonstrate that GLP-1Rs in the locus coeruleus (LC) are pharmacologically and physiologically relevant for food intake control. Furthermore, agonism of LC GLP-1Rs induces illness-like behaviors, and antagonism of LC GLP-1Rs can attenuate GLP-1R–mediated nausea. Electrophysiological and behavioral pharmacology data support a role for LC GLP-1Rs expressed on presynaptic glutamatergic terminals in the control of feeding and malaise. Collectively, our work establishes the LC as a site of action for GLP-1 signaling and extends our understanding of the GLP-1 signaling mechanism necessary for the development of improved obesity pharmacotherapies.
Article
Full-text available
Motions sickness (MS) occurs when the brain receives conflicting sensory signals from vestibular, visual and proprioceptive systems about a person’s ongoing position and/or motion in relation to space. MS is typified by symptoms such as nausea and emesis and implicates complex physiological aspects of sensations and sensorimotor reflexes. Use of animal models has been integral to unraveling the physiological causality of MS. The commonly used rodents (rat and mouse), albeit lacking vomiting reflex, reliably display phenotypic behaviors of pica (eating of non-nutritive substance) and conditioned taste aversion (CTAver) or avoidance (CTAvoi) which utilize neural substrates with pathways that cause gastrointestinal malaise akin to nausea/emesis. As such, rodent pica and CTAver/CTAvoi have been widely used as proxies for nausea/emesis in studies dealing with neural mechanisms of nausea/emesis and MS, as well as for evaluating therapeutics. This review presents the rationale and experimental evidence that support the use of pica and CTAver/CTAvoi as indices for nausea and emesis. Key experimental steps and cautions required when using rodent MS models are also discussed. Finally, future directions are suggested for studying MS with rodent pica and CTAver/CTAvoi models.
Article
Full-text available
Purpose: To assess the effectiveness and safety of acupuncture for the prevention of chemotherapy-induced nausea and vomiting (CINV), with a specific intention on exploring sources of between-study variation in treatment effects. Methods: MEDLINE, EMBASE, Cochrane CENTRAL, CINAHL, Chinese Biomedical Literature Database, VIP Chinese Science and Technology Periodicals Database, China National Knowledge Infrastructure, and Wanfang were searched to identify randomized controlled trials (RCTs) that compared acupuncture to sham acupuncture or usual care (UC). The main outcome is complete control (no vomiting episodes and/or no more than mild nausea) of CINV. GRADE approach was used to rate the certainty of evidence. Results: Thirty-eight RCTs with a total of 2503 patients were evaluated. Acupuncture in addition to UC may increase the complete control of acute vomiting (RR, 1.13; 95% CI, 1.02 to 1.25; 10 studies) and delayed vomiting (RR, 1.47; 95% CI, 1.07 to 2.00; 10 studies) when compared with UC only. No effects were found for all other review outcomes. The certainty of evidence was generally low or very low. None of the predefined moderators changed the overall findings, but in an exploratory moderator analysis we found that an adequate reporting of planned rescue antiemetics might decrease the effect size of complete control of acute vomiting (p = 0.035). Conclusion: Acupuncture in addition to usual care may increase the complete control of chemotherapy-induced acute vomiting and delayed vomiting but the certainty of evidence was very low. Well-designed RCTs with larger sample sizes, standardized treatment regimens, and core outcome measures are needed.
Article
Full-text available
Objective: To investigate the ability of preoperative neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) to predict postoperative nausea and vomiting (PONV) after total knee arthroplasty (TKA). Methods: The clinical data of 108 male patients with hemophilia A who underwent TKA an our institution were collected and analyzed. Confounding factors were adjusted by propensity score matching. The best cutoffs of the NLR and PLR were determined by the area under the receiver operating characteristic curve (ROC). The predictive ability of these indexes was assessed by measuring the sensitivity, specificity, and positive and negative likelihood ratios. Results: There were significant differences in the use of antiemetics (p = 0.036) and the incidence of nausea (p < 0.001) and vomiting (p = 0.006) between the two groups (NLR <2 and ≥2). An increase in preoperative NLR was an independent risk factor for PONV in patients with hemophilia A (p < 0.05). ROC analysis showed that NLR significantly predicted the occurrence of PONV (cutoff value: 2.20, ROC: 0.711, p < 0.001). In turn, the PLR did not strongly predict PONV. Conclusions: The NLR is an independent risk factor for PONV in patients with hemophilia A and can significantly predict this event. Thus, follow-up monitoring is essential for these patients.
Article
This review examines the role of the neuropeptide substance P within the neuroinflammation that follows traumatic brain injury. It examines it in reference to its preferential receptor; the neurokinin-1 receptor and explores the evidence for antagonism of this receptor in traumatic brain injury with therapeutic intent. Expression of substance P increases following traumatic brain injury. Subsequent binding to the neurokinin-1 receptor results in neurogenic inflammation, a cause of deleterious secondary effects that include an increased intracranial pressure and poor clinical outcome. In several animal models of TBI, neurokinin-1 receptor antagonism has been shown to reduce brain oedema and the resultant rise in intracranial pressure. A brief overview of the history of substance P is presented, alongside an exploration into the chemistry of the neuropeptide with a relevance to its functions within the central nervous system. This review summarises the scientific and clinical rationale for substance P antagonism as a promising therapy for human TBI.
Introduction: Post-operative nausea and vomiting (PONV) affects 30% of all patients undergoing surgery and up to 80% of high-risk patients. Antiemetics for PONV prophylaxis target a variety of receptor systems, with varying degrees of efficacy and side effect profile. Neurokinin -1 receptor antagonists are the most recent class of compounds investigated for PONV prophylaxis, with aprepitant being the only one currently approved for this indication. Areas covered: This review covers the pathophysiology of PONV, current recommendations for PONV prophylaxis, pharmacokinetics, and pharmacodynamics of aprepitant, and the evidence for its efficacy in the management of PONV as a single agent and in combination therapy. Expert opinion: Aprepitant is effective for PONV prophylaxis. It has superior antivomiting efficacy, long half-life, and favorable side effect profile. Data on antiemetic combinations involving aprepitant are limited, and it not clear if the addition of other antiemetics to aprepitant result in improved PONV prophylaxis. The oral route of administration of aprepitant is a potential limitation in a busy clinical practice. However, the recent approval of an intravenous formulation could provide a more convenient route of administration. Aprepitant remains more expensive than other antiemetics, and there are no studies assessing the cost effectiveness of its use.
Article
Full-text available
Guilt is a negative emotion, elicited by realizing one has caused actual or perceived harm to another person. Anecdotally, guilt often is described as a visceral and physical experience. However, while the way that the body responds to and contributes to emotions is well known in basic emotions, little is known about the characteristics of guilt as generated by the autonomic nervous system. This study investigated the physiologic signature associated with guilt in adults with no history of psychological or autonomic disorder. Healthy adults completed a novel task, including an initial questionnaire about their habits and attitudes, followed by videos designed to elicit guilt, as well as the comparison emotions of amusement, disgust, sadness, pride, and neutral. During the video task, participants’ swallowing rate, electrodermal activity, heart rate, respiration rate, and gastric activity rate were continuously recorded. Guilt was associated with alterations in gastric rhythms, electrodermal activity, and swallowing rate relative to some or all the comparison emotions. These findings suggest that there is a mixed pattern of sympathetic and parasympathetic activation during the experience of guilt. These results highlight potential therapeutic targets for modulation of guilt in neurologic and psychiatric disorders with deficient or elevated levels of guilt, such as frontotemporal dementia, posttraumatic stress disorder, and Obsessive-compulsive disorder.
Article
The selective NK 1 receptor antagonist, CP‐99,994, produced dose‐related (0.1‐1.0 mg kg ⁻¹ , s.c.) inhibition of vomiting and retching in ferrets challenged with central (loperamide and apomorphine), peripheral (CuSO 4 ) and mixed central and peripheral (ipecac, cisplatin) emetic stimuli. Parallel studies with the enantiomer, CP‐100,263 (1 mgkg ⁻¹ , s.c), which is > 1 000 fold less potent as a NK 1 antagonist, indicated that it was without significant effect against CuSO 4 , loperamide, cisplatin and apomorphine‐induced emesis. Against ipecac, it inhibited both retching and vomiting, expressing approximately l/10th the potency of CP‐99,994. The 5‐HT 3 receptor antagonist, tropisetron (lmgkg ⁻¹ , s.c.) inhibited retching and vomiting to cisplatin and ipecac, but not CuSO 4 or loperamide. CP‐99,994 (lmgkg ⁻¹ , i.v.) blocked retching induced by electrical stimulation of the ventral abdominal vagus without affecting the cardiovascular response, the apnoeic response to central vagal stimulation or the guarding and hypertensive response to stimulation of the greater splanchnic nerves. CP‐99,994 (1 mg kg ⁻¹ , i.v.) did not alter baseline cardiovascular and respiratory parameters and it failed to block the characteristic heart rate, blood pressure and respiratory rate/depth changes in response to i.v. 2‐methyl‐5‐HT challenge (von Bezold‐Jarisch reflex). Using in vitro autoradiography, [ ³ H]‐substance P was shown to bind to several regions of the ferret brainstem with the density of binding in the nucleus tractus solitarius being much greater than in the area postrema. This binding was displaced by CP‐99,994 in a concentration‐related manner. In dogs, CP‐99,994 (40 μg kg ⁻¹ bolus and 300 μg kg ⁻¹ h ⁻¹ , i.v.) produced statistically significant reductions in vomiting to CuSO 4 and apomorphine as well as retching to CuSO 4 . Together, these studies support the hypothesis that the NK 1 receptor antagonist properties of CP‐99,994 are responsible for its broad spectrum anti‐emetic effects. They also suggest that CP‐99,994 acts within the brainstem, most probably within the nucleus tractus solitarius although the involvement of the area postrema could not be excluded.
Article
The motor control of the lower esophageal sphincter (LES) is critical for normal swallowing and emesis, as well as for the prevention of gastroesophageal reflux. However, there are surprisingly few data on the central organization and neurochemistry of LES-projecting preganglionic neurons. There are no such data in ferrets, which are increasingly being used to study LES relaxation. Therefore, we determined the location of preganglionic neurons innervating the ferret LES, with special attention to their relationship with gastric fundus-projecting neurons. The neurochemistry of LES-projecting neurons was also investigated using two markers of “nontraditional” neurotransmitters in vagal preganglionic neurons, nitric oxide synthase (NOS), and dopamine (tyrosine hydroxylase: TH). Injection of cholera toxin B subunit (CTB)-horseradish peroxidase (HRP) into the muscular wall of the LES-labeled profiles throughout the rostrocaudal extent of the dorsal motor nucleus of the vagus (DMN) The relative numbers of profiles in three regions of the DMN from caudal to rostral are, 43 ± 5, 67 ± 11, and 113 ± 30). A similar rostrocaudal distribution occurred after injection into the gastric fundus. When CTB conjugated with different fluorescent tags was injected into the LES and fundus both labels were noted in 56 ± 3% of LES-labeled profiles overall. This finding suggests an extensive coinnervation of both regions by vagal motor neurons. There were significantly fewer LES-labeled profiles that innervated the antrum (16 ± 9%). In the rostral DMN, 15 ± 4% of LES-projecting neurons also contained NADPH-diaphorase activity; however, TH immunoreactivity was never identified in LES-projecting neurons. This finding suggests that NO, but not catecholamine (probably dopamine), is synthesized by a population of LES-projecting neurons. We conclude that there are striking similarities between LES- and fundic-projecting preganglionic neurons in terms of their organization in the DMN, presence of NOS activity and absence of TH immunoreactivity. Coinnervation of the LES and gastric fundus is logical, because the LES has similar functions to the fundus, which relaxes to accommodate food during ingestion and preceding emesis, but has quite different functions from the antrum, which provides mixing and propulsion of contents for gastric emptying. The presence of NOS in some LES-projecting neurons may contribute to LES relaxation, as it does in the case of fundic relaxation. The neurologic linkage of vagal fundic and LES relaxation may have clinical relevance, because it helps explain why motor disorders of the LES and fundus frequently occur together. J. Comp. Neurol. 430:222–234, 2001. © 2001 Wiley-Liss, Inc.
Article
We investigated involvement of the autonomic nervous system in gastric motor and cardiovascular responses to Δ9-tetrahydrocannabinol (Δ9-THC) in anesthetized rats. Intravenously administered Δ9-THC evoked long-lasting decreases in intragastric pressure and pyloric contractility, bradycardia, and hypotension. The changes in gastric motor function and bradycardia were abolished by vagotomy and ganglionic blockade, whereas spinal cord transection prevented the hypotensive response. Administered intravenously alone, N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide, a putative cannabinoid CB1 receptor antagonist, evoked transient decrease in intragastric pressure, and hypertension that was associated with bradycardia. However, this agent completely blocked the gastric motor and cardiovascular responses to intravenous Δ9-THC. Application of Δ9-THC to the dorsal surface of the medulla resulted in small and short-lasting decreases in gastric motor and cardiovascular function. We conclude that the decrease in gastric motor function and bradycardia are partially due to an action of Δ9-THC in the dorsal medulla and that intact vagal nerves are required. The hypotension was mediated through sympathetic pathways. Both gastric motor and cardiovascular effects of peripherally administered Δ9-THC seem to be mediated through cannabinoid CB1 receptors.
Article
The mechanisms underlying the frequent association of nausea and vomiting with elevations of plasma vasopressin(PAVP) were studied in man and rat. After oral water loads (N = 16), plasma osmolality fell in all human subjects and was associated with a decline in PAVP in 14 asymptomatic human subjects. In 2 human subjects, nausea occurred and was associated with increases in PAVP, without changes in blood pressure. During ethanol infusion (N = 28), PAVP was suppressed unless nausea supervened. In 4 nauseated human subjects, PAVP escaped from ethanol inhibition and rose to levels 10 times basal, despite the absence of hemodynamic changes. Apomorphine, a potent dopamine agonist and emetic agent, was administered to human volunteers in doses of 7 to 24 microgram/kg. There was no increase in PAVP in 3 human subjects who remained asymptomatic (7 to 16 microgram/kg). Ten human subjects experienced nausea after 16 microgram/kg, which was followed shortly by marked increases in PAVP. Emesis occurred in 5 human subjects given 16 to 24 microgram/kg, and was followed by PAVP levels similar to those seen with nausea alone. In 7 human subjects from the nausea group, the repeat study (16 microgram/kg) after pretreatment with dopamine antagonist (haloperidol, N = 4; fluphenazine, N = 3) resulted in complete blockage of apomorphine-induced AVP release. In rats, which lack an emetic reflex, apomorphine doses of 200 microgram/kg induced only slight increases in PAVP when compared to the response to 16 microgram/kg in man. These studies indicate that stimulation of the emetic reflex results in AVP-release in man. Nausea-mediated AVP release supervenes over concomitant osmolar or pharmacologic (ethanol) inhibition.
Article
This work was performed to prove the hypothesis that the pattern generator for the emetic act exists in the Bötzinger complex (BOT) and is driven by vagal afferents via the subpostrema portion of the nucleus of the solitary tract (mNST). Non-respiratory neurons (78) intermingling with BOT respiratory neurons in decerebrate dogs responded to pulse train stimulation of vagal afferents with a mean latency of 387 ms. During retching induced by vagal stimulation, one-half of the non-respiratory neurons exhibited high frequency burst firings synchronous with each retch (SH-firing, SH-neurons) and one-third of these neurons showed similar firings synchronous with the periods between retches (BH-firing, BH-neurons). Two-thirds of the SH-neurons and one-half of the BH-neurons fired with gradually augmenting frequencies (augmenting firing) during the period prior to retching, which may correspond to the period of prodromal signs of vomiting. Three SH-neurons were observed at fictive expulsion: all 3 exhibited burst firings concomitant with expulsion. During cooling block of transmission in the mNST, stimulation of the vagus nerve ipsilateral to the cooling failed to induce not only retching but also augmenting firing and SH-firing in all 11 BOT SH-neurons observed. In contrast, contralateral vagal stimulation induced retching and neuronal firings which had been observed before the cooling. These results support the hypothesis mentioned above. Respiratory firings were changed during retching in all BOT respiratory neurons observed. Respiratory firings were depressed during retching in the majority (15/25) of inspiratory (I) neurons and in a few expiratory (E) neurons (6/45). SH-firing was exhibited by 3 I- and 13 E-neurons. A few (2) I- and half (23) E-neurons showed BH-firing. These results indicate that all BOT respiratory neurons participate in central patterning of the emetic act.
Article
Roles of neurons in the nucleus of the solitary tract corresponding to the area subpostrema (mNST) for the retching reflex were investigated in decerebrate, paralyzed dogs. Retching was defined as rhythmic coactivation of the phrenic and abdominal muscle nerves. Retching which had been induced by stimulation of the left and right abdominal vagus nerves was impaired by cooling the left and right mNSTs, respectively. This result indicates that the mNST neurons mediate activities of emetic vagal afferents. All 40 non-respiratory neurons in the mNST, which had excitatory response to pulse train stimulation of the vagus nerve, were also activated by continuous stimulation of the vagus nerve to provoke retching. During provoked retching, however, these neurons did not exhibit any activities modulated in association with retching. The average latency of responses of these neurons to the pulse train stimulation (306.5 ms) was significantly shorter than that of the inspiratory neurons in the lateral NST and the adjacent reticular formation. Discharge frequencies of these neurons in the mNST gradually increased after administration of apomorphine (6/10) and glutamate (14/14) to the 4th ventricle. Antidromic responses to stimulation of the Bötzinger complex were observed in some (20/289) of the mNST neurons. These findings suggest that neurons in the mNST mediate the information from both the abdominal vagal afferents and the area postrema and drive the pattern generator for retching and vomiting, which is assumed to be located in the Bötzinger complex.
Article
Fundamental approaches in selection of new agents for evaluation in prevention of space/motion sickness (SMS) are reviewed. The discussion centers on drugs under investigation at the Johnson Space Center. Methodology that employs the rotating chair for measuring SMS symptomatology and susceptibility is described. The most obvious approach to the development of new agents relies on selection of agents from drug classes that possess pharmacologic properties of established anti-motion sickness agents. A second approach selects drugs that are used to prevent emesis caused by means other than exposure to motion. The third approach relies on basic research that characterizes individual differences in susceptibility. The hypothesis is: detection of individual differences leads to identification of specific drugs, which target physiologic systems that show individual differences. These physiologic systems are targets for therapy and may play a role in the etiology of SMS. Two drugs that reduce susceptibility to SMS include dexamethasone and d(CH2)5Tyr(Me)AVP, a vasopressin (AVP)V1 antagonist. The latter peptide has demonstrated complete blockade of emesis and other significant symptoms in squirrel monkeys. These studies were predicated on observations that subjects who were more resistant to SMS had higher plasma AVP after severe nausea than subjects with lower resistances. Investigations are underway to test a 0.5-mg intravenous dose in humans. Kappa opioid agonists inhibit AVP release and offer new therapeutic possibilities and advantages over AVP peptides. This review details the experimental data collected on AVP and adrenocorticotropin. The literature supports interrelated roles for AVP and opioid peptides in SMS. Experimental testing of kappa agonists is warranted because specific opioid agonists act at neuroanatomical sites causing nausea and vomiting. It is argued opioid receptors in the chemoreceptor trigger zone and vomiting center stimulate and inhibit the emetic response, respectively. The evidence suggests kappa and/or mu receptors at VC are involved in inhibition of emesis, whereas delta opioid receptors at CTZ are involved in stimulation of emesis.
Article
To clarify the location of the pattern generator for the emetic act, the bulb was systematically stimulated and partially cut in decerebrate, paralyzed dogs. Stimulation of the following bulbar structures elicited the activities which could be recognized as retching and vomiting in the following muscle nerves. The bulbar structures were: the intra-bulbar bundle of the vagal afferents, the solitary tract and the medial subdivision of its nucleus (NTS), the area postrema, the commissural nucleus, the raphe area at the obex level, and the longitudinal reticular column which consists of 3 areas--the area between the caudal parts of the solitary complex (SC) and the nucleus ambiguus, the area ventromedial to the rostral part of the nucleus and the area dorsomedial to the retrofacial nucleus (RFN) which may correspond to the Bötzinger complex (BOT). The muscle nerves were: the phrenic branches to the dome and hiatal parts of the diaphragm, the abdominal muscle nerve, the pharyngo-esophageal branch of the vagus nerve, the mylohyoid muscle nerve, and the recurrent nerve branches to the adductors and abductor of the glottis. Emetic responses to stimulation of the vagal ventral trunk and the rostral SC still remained after cutting of the bilateral SCs at about 1 mm rostral to the obex, but disappeared after cutting at about 3.5 mm rostral to the obex. After the rostral cuts, stimulation of the SC part caudal to the cuts and the reticular column still induced the emetic act. Emetic responses to stimulation of the caudal SC remained after transection of the bulb at the rostral end of the RFN, but disappeared after transection at its caudal end or after partial cutting of the caudal BOT. The following hypothesis was proposed from these results. Emetic vagal afferents enter the rostral bulb, then descend through the SC to the area subpostrema. Subpostrema neurons project through the reticular column to the pattern generator of the emetic act in the BOT and activate it.
Article
We have monitored the expression of c-fos protein in the medulla oblongata of the ferret, using immunocytochemistry, to identify the brainstem pathways involved in the mediation of nausea and vomiting caused by the antineoplastic drug cisplatin. Cisplatin administration resulted in c-fos-like immunoreactivity (FLI) in the area postrema, the nucleus of the solitary tract, and in scattered cells within the ependymal lining of the fourth ventricle. Unilateral cervical vagotomy greatly reduced FLI in the ipsilateral nucleus of the solitary tract but did not significantly affect reactivity in the contralateral solitary tract nucleus or in the area postrema. Pretreatment of the animals with the 5-HT3 antagonist granisetron (BRL 43694) abolished the retching and vomiting caused by cisplatin and markedly reduced the cisplatin-evoked FLI in the nucleus of the solitary tract; treatment with this drug had no significant effect on cisplatin-evoked FLI in the area postrema. The results suggest that cisplatin induces c-fos gene expression in the nucleus of the solitary tract by an action involving vagal afferent pathways and also by a vagally independent, direct action on the area postrema. The anti-emetic 5-HT3 antagonist drug granisetron mimicked the effect of vagotomy on c-fos protein induction suggesting that it may act via 5-HT3 receptors known to be associated with vagal afferent terminals. The FLI seen in the area postrema was neither vagally dependent nor was it abolished by granisectron.(ABSTRACT TRUNCATED AT 250 WORDS)