Sie sind auf Seite 1von 16

Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

www.elsevier.com/locate/autneu

Review

Signals for nausea and emesis: Implications for models


of upper gastrointestinal diseases
Paul L.R. Andrews a,*, Charles C. Horn b
a
Division of Basic Medical Sciences, St George’s University of London, Cranmer Terrace, London, SW 17 0RE, UK
b
Monell Chemical Senses Center, Philadelphia, PA 19104, USA

Received 14 January 2006; received in revised form 14 January 2006; accepted 14 January 2006

Abstract

Nausea and vomiting are amongst the most common symptoms encountered in medicine as either symptoms of diseases or side effects of
treatments. In a more biological setting they are also important components of an organism’s defences against ingested toxins. Identification
of treatments for nausea and vomiting and reduction of emetic liability of new therapies has largely relied on the use of animal models, and
although such models have proven invaluable in identification of the anti-emetic effects of both 5-hydroxytryptamine3 and neurokinin1
receptor antagonists selection of appropriate models is still a matter of debate. The present paper focuses on a number of controversial issues
and gaps in our knowledge in the study of the physiology of nausea and vomiting including: The choice of species for the study of emesis and
the underlying behavioural (e.g. neophobia), anatomical (e.g. elongated, narrow abdominal oesophagus with reduced ability to shorten) and
physiological (e.g. brainstem circuitry) mechanisms that explain the lack of a vomiting reflex in certain species (e.g. rats); The choice of
response to measure (emesis[retching and vomiting], conditioned flavour avoidance or aversion, ingestion of clay[pica], plasma hormone
levels[e.g. vasopressin], gastric dysrhythmias) and the relationship of these responses to those observed in humans and especially to the
sensation of nausea; The stimulus coding of nausea and emesis by abdominal visceral afferents and especially the vagus—how do the
afferents encode information for normal postprandial sensations, nausea and finally vomiting?; Understanding the central processing of
signals for nausea and vomiting is particularly problematic in the light of observations that vomiting is more readily amenable to
pharmacological treatment than is nausea, despite the assumption that nausea represents ‘‘low’’ intensity activation of pathways that can
evoke vomiting when stimulated more intensely.
D 2006 Elsevier B.V. All rights reserved.

Keywords: Nausea; Vomiting; Vagus; Pica; Flavour aversion; Conditioned taste aversion

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
2. Choice of species in nausea and emesis research . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
2.1. The vomiting reflex is lacking in some species . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
2.2. What do conditioned flavour avoidance (CFA) and clay ingestion (pica) measure?. . . . . . . . . . . . . . . . . . . 103
2.2.1. Conditioned flavour avoidance (CFA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
2.2.2. Kaolin ingestion (pica) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
2.3. Neurohypophyseal hormonal secretions and gastric dysrhythmias as measures of nausea? . . . . . . . . . . . . . . . 107
3. The role of pain in nausea and vomiting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108

* Corresponding author.
E-mail address: pandrews@sgul.ac.uk (P.L.R. Andrews).

1566-0702/$ - see front matter D 2006 Elsevier B.V. All rights reserved.
doi:10.1016/j.autneu.2006.01.008
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 101

4. Neural circuitry for nausea . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108


4.1. Visceral afferent fibres . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
4.2. Direct action of stimuli on the brain? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
4.3. The stimulus and coding of signals for nausea and vomiting. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110

1. Introduction 2. Choice of species in nausea and emesis research

Nausea and vomiting are amongst the most common Vertebrates (fish, amphibia, reptiles, birds and mammals)
symptoms encountered in medicine. They occur separately have significant species differences in the mechanisms and
and together in a diverse range of diseases, following especially the mechanics of vomiting that make it difficult to
anaesthesia and surgery, and are side effects of both current make more than general comparisons between animals. Even
drug treatments and novel therapies in development where amongst the mammals it may be difficult to extrapolate
there may be dose limiting toxicities (for review, see Rudd findings from laboratory animal studies to explain nausea and
and Andrews, 2005). Nausea and vomiting are also vomiting in humans, especially as we begin to understand
components of the body’s defensive response to toxins more of the species differences between neurotransmitter
accidentally ingested with food. Understanding the pathways receptors and the way that they interact with synthetic ligands
by which the sensation of nausea is generated and reflex (e.g., neurokinin NK1 receptors; Andrews and Rudd, 2004).
vomiting is evoked is essential to identifying novel Emesis (retching and vomiting), which can be overtly
pharmacological approaches to the development of anti- measured in some laboratory animals, is much easier to
emetics and also for gaining insights into the underlying study and interpret than behaviours that have been associated
defect in disorders where nausea and vomiting are cardinal with the subjective experience of nausea. However, even with
features, such as cyclic vomiting syndrome where patients research on vomiting there are important species differences.
may have a median peak intensity of 6 vomits an hour at the
height of an attack (Li and Fleisher, 1999). The investigation 2.1. The vomiting reflex is lacking in some species
of pathways and their pharmacology relies to a large extent
upon the use of appropriate animal models and this paper In general, the vomiting reflex is arguably not essential
will focus on some of the issues related to the selection of for survival; but it is advantageous for ridding the body of
models for the investigation of nausea and vomiting and also ingested toxins and may prove particularly useful for a
on the limitations of using species without an emetic reflex species in a specific niche. Several mammalian species
(e.g. rats) to study malaise originating in the upper gut. commonly used in laboratory research, including the rat,
In addition, research into nausea and vomiting provides mouse, hamster, guinea pig, and rabbit, appear not to have
important insights into information processing in the a vomiting reflex. Although a large number of toxicosis
autonomic nervous system. For example, the abdominal studies have been conducted in the rat, a relatively limited
vagal afferent fibres can evoke both responses, the nucleus number of stimuli, including, gastrointestinal irritants,
tractus solitarius (NTS) plays a major role in integrating the motion, apomorphine, and cytotoxic drugs, have been
emetic response and is a target for anti-emetic agents, and used to study behaviours associated with malaise. While
the autonomic efferents mediate many of the physiological there is little doubt that overt emesis has not been observed
changes that accompany both nausea (e.g., cutaneous in the laboratory rat, only a few strains (e.g., Wistar) have
vasoconstriction and sweating) and vomiting (e.g., proxi- been studied, and this may be significant because it is
mal stomach relaxation, retrograde giant contraction of the known that conditioned flavour avoidances are variably
small intestine). Furthermore, because some motor compo- induced in different strains of mice (e.g., Risinger and
nents (e.g. inhibition of the crural diaphragm) of the emetic Brown, 1996; Risinger and Cunningham, 2000; Bielavska
reflex occur in other reflexes, such as belching, and et al., 2002). Furthermore, there are isolated reports of
diseases, for example gastro-oesophageal reflux disease, ‘‘retching’’ but not vomiting in mice, which if replicated
the study of emesis may provide fortuitous insights into the may suggest that they have a degenerate reflex rather than
pathophysiology and treatment of other disorders. In this an absent one (e.g., Furukawa and Yamada, 1980). Rats do
context it is important to note that several ‘‘agonist’’ anti- have a gag reflex triggered by mechanical stimulation of
emetics are also effective in gastroesophageal reflux disease the pharynx (Andrew, 1956) and a gag has features that
or models (e.g., the opioid receptor agonist morphine; could be considered similar to a single retch. This
cannabinoid CB1 receptor agonists, GABA B receptor observation provides some evidence that the emetic reflex
agonists (Penagini and Bianchi, 1997; Holloway, 2001; is degenerate in rodents. Some potentially emetic stimuli
Tonini et al., 2004). have been used in mice, hamsters, and rabbits to induce
102 P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

conditioned flavour avoidance or pica (measures of motor neurons, that might be important for the production of
malaise; see below) and have failed to induce emesis emesis appear to be absent in the rat compared to the ferret,
(e.g., Hobbs et al., 1976; Fox, 1977; Santucci et al., 2000; an emetic species (Dobbins and Feldman, 1994; Yates et al.,
Christian et al., 2001; Santucci et al., 2002; Yamamoto et 1999). It is worth noting that rats, like emetic species, have
al., 2002a). However, before stating that all rodents and an area postrema (AP) but it is difficult to assign specific
lagomorphs lack an emetic reflex it may be wise to functions to this brain region because lesions of this site
undertake further formal studies of this phenomenon using ablate vagal sensory neurons that terminate in the AP and
a wide range of doses and stimuli. As an alternative to often destroys some portion of the adjacent NTS, which
emesis, the rat and other non-vomiting species probably receives a large vagal sensory projection from the gut
use rapidly learned aversions to avoid toxicosis (see (Norgren and Smith, 1988). The AP appears to be non-
below). This is supported by the ‘‘nibbling’’ type of food essential for the production of vomiting, e.g., motion can
intake in these species and some indications that they are produce emesis in the cat and monkey after AP ablation
neophobic (avoiding novel foods or tasting a small amount (Fox et al., 1990) and gastric irritants (e.g., copper sulphate)
and returning later to ingest more if illness does not occur) can also induce emesis in the absence of the AP in the dog
(Mitchell, 1976). Both behaviours would serve to limit the (Andrews et al., 2001). Other reflex neural circuitry of the
consumption of a potentially poisonous meal. Furthermore, brainstem appears to be similar between vomiting and non-
the rat and mouse possess more hepatic genes involved in vomiting species, e.g., the cardio-respiratory reflexes in rat
detoxification than the human, and therefore the emetic versus Suncus (Paton et al., 2000; Smith et al., 2001b). It
reflex might provide less advantage in these species (Gibbs should also be noted that although rats do not vomit to intra-
et al., 2004). The Rodentia are a very successful Order gastric stimuli that evoke emesis in susceptible species (e.g.
adapting to a variety of habitats and they account for ferret) (Andrews et al., 1990), rats do respond to stimuli
> 40% of all mammalian species (Dawkins, 2004), which such as veratridine and high concentrations of KCl and
provides support for the comment that the vomiting reflex NaCl by markedly delaying gastric emptying (see Fig. 1). A
is not essential for survival. delay in gastric emptying has been argued to be a
Although apparently absent in rodents and lagomorphs, component of the defensive response in rodents and in
the vomiting reflex is present in representatives of several emetic species because this action would delay delivery of
mammalian Orders, including Primates (e.g. human, Cyn- any toxin into the small intestine where it may produce more
omologous monkey, marmoset), Carnivora (dog, cat, ferret), damage, especially if absorbed (Davis et al., 1986).
Cetartiodactyla (e.g. pig, sperm whale), and Insectivora (e.g. Among those animals that have a vomiting reflex there
house musk shrew, Suncus murinus), and it is also present are several important issues that affect the interpretation of
throughout the Vertebrates, being demonstrated in proto- research findings across species. (1) There is no consistent
typical fish, amphibia, reptiles and birds (Andrews et al., profile across species for the rank order of sensitivity to
1990, 2000; King, 1990; Sims et al., 2000; Tanihata et al., different emetic stimuli. Although data are relatively limited
2004). A discussion of the vomiting mechanisms in these in some species it is clear that the sensitivity profile to emetic
non-mammalian species is beyond the scope of the current
paper.
There are anatomical and neural circuitry differences 6
Fluid in stomach (ml) after 1 h

between vomiting and non-vomiting mammalian species


that would make vomiting difficult for non-emetic species 5 *** **
even if they attempted to. The abdominal oesophagus is
4
***
relatively wider and shorter in emetic species (Andrews,
1995). In addition, a comparison of the mouse and Suncus
3
revealed that in the non-vomiting mouse the vagus had little
ability to shorten the oesophagus longitudinally and hence 2
reduce one of the resistances to the flow of vomitus
(Andrews et al., 2003). In general, the diaphragm appears 1
to be more muscular in the emetic species so far studied
with a reduced area of central tendon but this is not a 0
universal finding (Andrews, unpublished observations) and
e

e
aC

C
lin

in
K

id
N
Sa

the non-emetic roles of the diaphragm (e.g. breathing,


M

tr
M

ra
1
1

coughing, sneezing, belching, hiccupping) make the signif-


Ve

icance of this observation difficult to link to emesis. Taken


Fig. 1. Effect of potentially emetic solutions on gastric emptying in the
overall these structural characteristics probably provide
rat. Five millilitres of each solution was gavaged into the stomachs of
greater propulsive power and less resistance for expelling rats previously deprived of food overnight (n = 5). ***p < 0.0001 versus
vomitus. Furthermore, the brainstem circuitry, e.g., medial saline and **p < 0.001 versus saline. (Andrews, Payne and Hawthorne,
medullary reticular neurons that provide input to phrenic unpublished)
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 103

stimuli is species dependent. For example, amongst species by which animals deal with toxicosis. Furthermore, unlike
sensitive to apomorphine the rank order of emetic sensitivity emesis, we lack an adequate understanding of the physiol-
is dog > human > ferret >cat (note: the cat requires doses in ogy of nausea and have little basis for comparing the
the mg/kg range in comparison to other species where the behaviours of CFA and pica with reports of nausea in
dose is Ag/kg), with Suncus and monkey being unresponsive. humans. However, investigations of the neural systems that
For the chemotherapeutic agent cisplatin the rank order is produce CFA and pica might shed some light on the
monkey > human > dog > ferret > cat > Suncus; and for total physiology of human nausea because these systems appear
body X-radiation ferret > dog > human = monkey > cat to have related neural circuitry and pharmacology, e.g., anti-
(Andrews et al., 1990). Furthermore, the dog is sensitive to emetic drugs inhibit CFA and pica (e.g., Limebeer and
swinging-type motion whereas the cat is resistant (Noble, Parker, 2000; Saeki et al., 2001; Rudd et al., 2002). Table 1
1945) and it is argued that dogs appear to have approxi- shows a list of species and stimuli that have been using to
mately the same sensitivity to motion sickness as man study emesis, CFA, and pica.
(Money, 1970). Even amongst monkeys there are reports of
sensitivity differences between squirrel and rhesus monkeys 2.2.1. Conditioned flavour avoidance (CFA)
(Corcoran et al., 1990). (2) Not all emetic species are Many species show CFA or learned flavour aversion to
sensitive to all stimuli that cause vomiting. This is illustrated foods or flavours associated with toxicosis, including
by Suncus, which unlike most vomiting species does not humans (Mattes et al., 1991; Mattes, 1994). CFA is used
show emesis to morphine and apomorphine treatments extensively to study malaise in laboratory animals, partic-
(Selve et al., 1994). Apomorphine-induced emesis, although ularly the rat. In many cases this effect is simply called a
present in humans, is reported to be absent in the monkey conditioned ‘‘taste’’ aversion or ‘‘CTA.’’ However, there are
(Brizzee et al., 1955) but present in another primate, the two problems with this terminology. First, the modality of
marmoset (Costall et al., 1987). (3) In some cases the the conditioned stimulus is not always known, particularly
function of emesis is different. In most animals vomiting is with learned responses to complex foods. The conditioned
a protective function that serves to expel toxins from the stimulus could be any one of the three components of
gastrointestinal tract prior to absorption, but in some flavour, which include gustation, olfaction, or chemaesthe-
species it is also used to rid the gut of indigestible material, sia (touch, temperature, or irritation sensitivity, subserved by
such as bones from the guts of birds, hairballs from trigeminal innervation of the oral cavity), or a combination
crocodiles and indigestible squid beaks in sharks (Darolova, of these. Therefore the phenomenon is more properly
1991; Andrews et al., 2000), whilst vomiting is used as referred to as a conditioned ‘‘flavour’’ avoidance or aversion
means of defence when Petrels are threatened. In addition, in a general sense, particularly when the modality of the
some species regurgitate partially digested food from the flavour component for the conditioned stimulus has not
stomach to feed the young and relatives (e.g. wild dogs; been determined.
Van Lawick-Goodall and Van Lawick-Goodall, 1970). The second problem is the distinction between condi-
Emesis might also serve as an anticipatory response tioned flavour avoidance and aversion. Typically in testing
generated by cognitive, visual, or flavour stimuli, and this phenomenon in laboratory animals, a novel flavour is
several categories of stimuli produce incidental activation paired with injection of a suspected toxin. When animals are
of the emetic reflex, including chemotherapy, stress, intra- later tested they avoid consumption of a flavoured solution
cranial pressure, and motion. Other behaviours also appear that was associated with toxicosis, demonstrating a CFA. In
to be very similar to emesis. Although rumination and contrast, a conditioned flavour ‘‘aversion’’ must be tested by
regurgitation are functionally different from vomiting and measuring consumption of a flavoured fluid that is directly
lack the propulsive power of emesis, the neural systems infused through a cannula implanted in the oral cavity
engaged in these different behaviours might be similar, e.g., (Parker, 1995). This methodological difference can critically
in sheep (Carr et al., 1983). affect the interpretation of results because even drugs with
rewarding properties, such as amphetamines and cocaine,
2.2. What do conditioned flavour avoidance (CFA) and clay produce CFA, but do not generate learned flavour aversions
ingestion (pica) measure? in the rat (Parker, 1995); however, there may be important
species differences here because some emetic species show
In species that do not possess a vomiting reflex, such as amphetamine-induced CFA, e.g., cat and ferret, but Suncus
the rat, it is common to measure a conditioned flavour does not (Rabin and Hunt, 1992). Consequently, CFA may
avoidance (CFA) or clay ingestion (pica; consumption of not always represent malaise in the rat. For example, an anti-
non-nutritive substances) as an index of malaise. It is emetic drug, a 5-HT3 (5-hydroxytryptamine; serotonin)
tempting to be anthropomorphic and conclude that these receptor antagonist, had no effect on CFA but did attenuate
measures reflect ‘‘nausea,’’ but we have no way of knowing learned flavour aversions (tested with intra-oral infusions)
whether this is true with reference to the human experience produced by intra-peritoneal injections of lithium chloride
of nausea. We know that CFA and pica are associated with (LiCl, a gastrointestinal irritant) (Rudd et al., 1998;
sickness and malaise, and appear to be additional strategies Limebeer and Parker, 2000). Nearly all of the references
104 P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

Table 1
Mammalian species and selected stimuli used to elicit emesis, conditioned flavour avoidance (CFA), and picaa
Emesis CFA Pica
Cat Cytotoxic drugs: Cisplatin (McCarthy and Borison,
1984),
Cyclophosphamide (Fetting et al.,
1982)
Intragastric irritants: Copper sulfate (Kayashima and
Hyama, 1976)
Motion (Crampton and Daunton, 1983) (Fox et al., 1990)
Apomorphine (Costello and Borison, 1977)
Morphine (Villablanca et al., 1984)
Radiation (Rabin et al., 1986b) (Rabin et al., 1986b)
Nicotine (Beleslin et al., 1981)
Hormones and Angiotensin II (Rabin et al.,
neurotransmitters: 1986a)
Other: Amphetamine
(Rabin and Hunt, 1992)
Dog Cytotoxic drugs: Cisplatin (Gylys et al., 1979),
Cyclophosphamide (Amber et al.,
1990)
Intragastric irritants: Ipecac (Gardner et al., 1996), LiCl (Vavilova and Kassil, 1984)
Copper sulfate
(Kayashima and Hayama, 1975)
Apomorphine (Harrison et al., 1972)
Morphine (Lefebvre et al., 1981)
Radiation (Cooper and Mattsson, 1979)
Nicotine (Vig, 1990)
Hormones and CCK (Levine et al., 1984),
neurotransmitters: Vasopressin (Wu et al., 1985),
Peptide YY (Smith et al., 1989)
Ferret Cytotoxic drugs: Cisplatin, cyclophosphamide
(Andrews et al., 1990)
Intragastric irritants: Copper sulfate, ipecac, NaCl, KCl LiCl (Rabin and Hunt, 1992)
(Andrews et al., 1990)
Apomorphine (Andrews et al., 1990)
Morphine (Barnes et al., 1991)
Radiation (Andrews et al., 1990)
Guinea pig Intragastric irritants: LiCl (Braveman, 1974)
Hamster Cytotoxic drugs: Cyclophosphamide
(Hobbs et al., 1976)
Intragastric irritants: LiCl (Fox, 1977)
Apomorphine (Nowlis et al., 1980)
Nicotine (Etscorn et al., 1986)
Other: 2-deoxy-d-glucose
(Dibattista, 1988)
Human Cytotoxic drugs: Chemotherapy (Rudd and Andrews, Chemotherapy (Schwartz et al.,
2005) 1996)
Intragastric irritants: Ipecac (Jackson and Smith, 1978), Ipecac (Jackson and Smith, 1978)
Copper sulfate (Liu et al., 2001)
Motion (Yates et al., 1998) (Arwas et al., 1989)
Apomorphine (Schofferman, 1976)
Morphine (Bailey et al., 1993)
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 105

Table 1 (continued)
Emesis CFA Pica
Human Radiation (Cordts et al., 1987) (Carrell et al., 1986)
Hormones and CCK (Miaskiewicz et al., 1989)
neurotransmitters:
Other: Pregnancy (Weigel and Weigel, Pregnancy (Bayley et al., 2002) Pregnancy (Corbett et al.,
1989), 2003; Lopez et al., 2004),
Reduced intracranial pressure Gastric bypass surgery
(Mokri, 2004) (Kushner et al., 2004)
Monkey Cytotoxic drugs: Cisplatin (Fukui et al., 1993) Cyclophosphamide
(Hikami et al., 1990)
Intragastric irritants: Copper sulfate (Fukui et al., 1993) LiCl (Bergman and Glowa, 1986)
Motion (Wilpizeski et al., 1987) (Wilpizeski et al., 1987)
Radiation (Brizzee, 1956)
Nicotine (Spealman, 1983)
Hormones and CCK (Perera et al., 1993)
neurotransmitters:
Mouse Cytotoxic drugs: Cisplatin (Yamamoto
et al., 2002a)
Intragastric irritants: LiCl (Risinger and
Cunningham, 2000)
Motion (Kinney et al., 1993) (Santucci et al., 2002)
Radiation (Kimeldorf et al., 1960)
Nicotine (Etscorn, 1980)
Other: Magnetic field (Lockwood
et al., 2003)
Rat Cytotoxic drugs: Cisplatin (Rudd et al., 1998), Cisplatin (Takeda et al.,
Cyclophosphamide (Ader, 1976) 1993)
Intragastric irritants: Ipecac (Rudd et al., 1998), LiCl, Copper sulfate
Copper sulfate (Coil et al., (Hasegawa et al., 1992)
1978), LiCl (Coil et al., 1978)
Motion (Hutchison, Jr., 1973) (Mitchell et al., 1977a)
Apomorphine (Krane et al., 1976) (Hasegawa et al., 1992)
Morphine (Aung et al., 2004)
Radiation (Garcia et al., 1955) (Yamamoto et al., 2002b)
Nicotine (Etscorn et al., 1987)
Hormones and NPY (Sipols et al., 1992), NPY (Woods et al.,
neurotransmitters: CCK (Ervin et al., 1995) 1998),
CCK (McCutcheon et al.,
1992)
Other: Fat oxidation inhibitors (Singer 2-deoxy-d-glucose
et al., 1999), 2-deoxy-d-glucose (Watson et al., 1987)
(Stephan et al., 1999), Magnetic
field (Houpt et al., 2003)
House Musk Cytotoxic drugs: Cisplatin, cyclophosphamide
Shrew (Matsuki et al., 1988)
(Suncus Intragastric irritants: Copper sulfate, ipecac (Ueno et al., LiCl (Smith et al., 2001a)
murinus) 1987), LiCl (Parker et al., 2004)
Motion (Ueno et al., 1988) (Smith et al., 2001a)
Radiation (Torii et al., 1993)
Nicotine (Ueno et al., 1987) Smith et al., 2001a)
Hormones and Vasopressin (Ikegaya and Matsuki,
neurotransmitters: 2002)
a
For some stimuli only representative studies or review papers are cited. Only the most common mammalian species used in the study of emesis and malaise
are shown. A common set of emetic and nauseogenic stimuli, including cytotoxic drugs, intragastric irritants, etc., are included, but a few less commonly used
stimuli are also listed because they demonstrate the broad spectrum of stimuli that can produce emesis, CFA, and pica.
106 P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

cited in the present review refer to results with bottle generate learned flavour aversions (Sipols et al., 1992),
consumption measures, and are therefore tests of CFA and which suggests that the hunger stimulus can also produce
may not represent learned flavour aversions. conditioned flavour aversion.
It is likely that learned flavour aversion is an indicator
of malaise or nausea, and this type of learning might serve 2.2.2. Kaolin ingestion (pica)
to predict foods that should be avoided. A similar learning Another way to test for malaise is to measure the
process may occur in cancer patients because they often consumption of non-nutritive substances following a toxic
show learned avoidance to foods, or even environmental treatment. Ingestion of dirt or clay, also called pica, in
stimuli, associated with chemotherapy (Mattes et al., 1991; response to toxicosis is a common phenomenon in animals
Mattes, 1994; Matteson et al., 2002). In laboratory animals and is also observed in humans (Reid, 1992; Root-
learned flavour aversion produced by emetogenic treat- Bernstein and Root-Bernstein, 2000; Engel, 2002; Kushner
ments, including cytotoxic chemotherapy agents, can be et al., 2004). Rats injected with toxins or subjected to
blocked by anti-emetic medications. Ondansetron, a 5-HT3 motion consume non-nutritive clay (kaolin) that they
receptor antagonist, and delta-9-tetrahydrocannabinol, a would normally not ingest (Mitchell et al., 1977a; Takeda
cannabinoid receptor agonist, which inhibit nausea and et al., 1993; Santucci et al., 2000; Saeki et al., 2001;
vomiting in humans (e.g., Cassidy et al., 1988; Massidda Yamamoto et al., 2002a). The presence of pica in mice is
and Ionta, 1996), suppress learned flavour aversions in rats controversial with some studies reporting it (Santucci et
produced by intra-peritoneal injections of LiCl or cyclo- al., 2000, 2002; Yamamoto et al., 2002a) whilst others do
phosphamide, a chemotherapy agent (Limebeer and Parker, not (Liu et al., 2005). The consumption of clay induced by
1999, 2000). toxicosis might represent an adaptive response to bind or
The relation between CFA or learned flavour aversion dilute a toxin in the gastrointestinal tract and reduce its
and malaise might be directly assessed by using emeto- adverse effects on the organism. Clay is particularly
genic stimuli to produce CFA in emetic species. To date, effective for binding and diluting toxic chemicals (e.g.,
there are no reports showing conditioned flavour aversion, Phillips et al., 1995; Sarr et al., 1995; Phillips, 1999), and
using intra-oral infusion testing, in an emetic species. pica seems to be a specific response in rats because they
However, several studies show toxicosis-induced CFA in select clay rather than pebbles or soil when poisoned
emetic species, including human, cat, dog, monkey, (Mitchell et al., 1976). However, pica can also be related
Suncus, and ferret (e.g., Jackson and Smith, 1978; to nutritional needs; mineral deficiencies in humans and
Vavilova and Kassil, 1984; Rabin et al., 1986b; Fox et other animals can promote ingestion of clay and other
al., 1990; Rabin and Hunt, 1992; Smith et al., 2001a). materials that are rich in minerals (e.g., Reynolds et al.,
These studies suggest a complex relation between a toxin’s 1968; Smith and Halsted, 1970; Roselle, 1970).
emetogenic potential and its potency to produce a CFA. Evidence indicates that the positive relation between
For example, although radiation and LiCl produce vomit- level of toxicosis and pica is not always maintained at
ing in the ferret, LiCl treatment, but not radiation, induced higher doses of a toxin. For example, a high dose of
a CFA (Rabin and Hunt, 1992). Further studies using cisplatin, 6 mg/kg, in the rat failed to generate the delayed
conditioned flavour aversion testing, with intra-oral infu- phase of kaolin consumption, 48 –72 h after injection, that
sion testing, might clarify the relation between emesis, was characteristic of a lower dose, 3 mg/kg; and the high
learned flavour aversion, and CFA. dose was associated with successive reductions in food
It is difficult to assess the physiology for the generation intake and body weight over the 3 days after cisplatin
of malaise using CFA or learned flavour aversion testing treatment (Rudd et al., 2002). This effect appears to be
because many pharmacological and lesion treatments to the counterintuitive but high doses of toxins might make
brain affect neural processes, such as learning and memory, animals unable to eat kaolin. However, this phenomenon
that are not specifically involved in the systems for toxin needs to be investigated using stimuli other than cisplatin as
detection. Additionally, it is very difficult to assess the it induces marked gastric stasis and the reduction in kaolin
properties of the toxin detection system using CFA testing intake may be secondary to this effect.
because the conditioning protocol involves a delay in testing There are compelling relations between pica, CFA and
that is not conducive to a detailed temporal analysis of emesis. Like CFA, pica can be a learned response.
malaise. For example, it is clear from research on emesis Treatment with LiCl or cyclophosphamide produce CFA
that an analysis of the acute and delayed phases of toxin to a saccharin flavoured solution in the rat and subsequent
detection is critical, but this type of investigation is not access to this flavour will stimulate kaolin intake (Mitchell
possible using CFA or learned flavour aversion testing et al., 1977b). Furthermore, there appears to be a close
(Hesketh et al., 2003). Although CFA and learned flavour association between the neuropharmacology of toxicosis-
aversion appear to measure the malaise producing potential induced pica and emesis. Like emesis in cancer patients
of a stimulus, this is probably not always true. For example, receiving chemotherapy (Tsukada et al., 2001; Hesketh et
intra-cerebroventricular infusions of neuropeptide Y pro- al., 2003; de Wit et al., 2004), cisplatin-induced pica in
duce robust increases in food intake in the rat, but also the rat can be characterized into acute and delayed phases
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 107

that are most sensitive to 5-HT3 and NK1 receptor be particularly likely in the case of the circular vection
antagonists, respectively (Saeki et al., 2001; Rudd et al., model of motion sickness (Kim et al., 1997).
2002). However, the relation between emesis and pica is Animal models also show an increase in neurohypophy-
not always so clear. Diphenidol, an anti-emetic drug in seal hormonal secretions after treatments that produce
humans, is a potent inhibitor of apomorphine-induced pica emesis or malaise. Doses of apomorphine and cholecysto-
in the rat (Takeda et al., 1995), but does not affect emesis kinin (CCK) that induce emesis in the ferret also elevate
induced by apomorphine in the dog or ferret (Nakayama plasma vasopressin (Hawthorn et al., 1988; Billig et al.,
et al., 2004). To directly relate pica to emesis it is 2001), and vasopressin is elevated by electrical stimulation
important to test both phenomena in the same species. of abdominal vagal afferent fibres (Hawthorn et al., 1988).
Two studies using Suncus have addressed this issue and Importantly, it is plasma oxytocin levels, much more than
show that although emesis was induced by treatment with vasopressin, that is elevated following malaise producing
copper sulphate, nicotine, and cisplatin, none of these treatments in the rat, e.g., LiCl and CCK treatments
agents stimulated pica (Yamamoto et al., 2004; Liu et al., (Flanagan et al., 1988; Huang et al., 2000). In contrast
2005). emetigenic doses of apomorphine and CCK did not produce
an increase in plasma oxytocin levels in the ferret
2.3. Neurohypophyseal hormonal secretions and gastric (Hawthorn et al., 1988; Billig et al., 2001). These differ-
dysrhythmias as measures of nausea? ences in toxicosis induced neurohypophyseal secretions
between species needs to be further studied in other emetic
Toxins and treatments that produce nausea and vomiting and non-emetic species. In the rat, elevated oxytocin has
can potentially inhibit behavioural systems that generate been implicated in the mechanisms by which they cope with
CFA and pica, which might lead to an underestimation of stress induced by non-noxious sensory stimuli, whereas
the level of malaise. One way to avoid this problem is to noxious stimuli in the rat are associated with corticotrophin-
measure a correlate of malaise that does not depend on releasing factor (CRF) and vasopressin secretion (Uvnas-
animal behaviour. Blood levels of the neurohypophyseal Moberg, 1998).
hormones, vasopressin (ADH) and oxytocin, and gastric Despite a focus on vasopressin secretion as a correlate of
dysrhythmia are two physiological parameters that are nausea (and oxytocin as a correlate of malaise in the rat)
proposed to be correlates of malaise in animal models and other hormones are increased in subjects with nausea
reports of nausea in humans. induced by apomorphine, ipecac, and motion, including
Although vasopressin is normally secreted in response to adrenaline, cortisol, growth hormone, prolactin, adrenocor-
reduced hydration, vasopressin is elevated in well hydrated ticotrophic hormone, and pancreatic polypeptide (Ever-
humans who experience nausea, e.g., nausea produced by smann et al., 1978; Feldman et al., 1988; Nussey et al.,
gastric distension with a water load can elevate vasopressin 1988; Page et al., 1990; Xu et al., 1993). It remains to be
levels (Rowe et al., 1979). Many treatments that produce determined whether any of these hormones are related to the
nausea and vomiting, such as apomorphine, chemotherapy, genesis for the sensation of nausea in contrast to being
motion, and cholecystokinin, also increase plasma vaso- indicators of the generally ‘‘stressful’’ nature of malaise. It
pressin to varying extents including to levels exceeding would be helpful to have a plasma ‘‘endocrine profile’’
maximal levels for anti-diuresis (Rowe et al., 1979; Fisher et monitoring a wide range of signalling molecules with
al., 1982; Grant et al., 1986; Feldman et al., 1988; multiple emetic challenges under carefully controlled
Miaskiewicz et al., 1989; Edwards et al., 1989; Koch et conditions to identify consistent directional changes associ-
al., 1990b). Plasma vasopressin levels appear to increase ated with nausea (cf. gene microarray approach).
slightly before or at the onset of nausea (Miaskiewicz et al., A second important correlate of nausea is gastric
1989; Koch et al., 1990b). Administration of vasopressin at dysrhythmia, which is an abnormal frequency of the
supraphysiological doses produced nausea in humans, but myoelectric activity of the stomach. Motion or intravenous
when vasopressin was infused to match the plasma level infusion of vasopressin elicited gastric dysrhythmia in
during nausea induced by motion nausea was not reported humans, and the dysrhythmia was recording prior to
(Kim et al., 1997). Additionally, apomorphine induces reported nausea (Stern et al., 1987; Koch et al., 1990a;
nausea in patients with idiopathic diabetes insipidus despite Caras et al., 1997). Gastric dysrhythmia is closely correlated
the absence of an increase in plasma vasopressin (Nussey et with reported nausea in a variety of conditions that produce
al., 1988). Although this does not exclude a role for nausea, including surgery, pregnancy and chemotherapy
vasopressin in the genesis of nausea in intact individuals, (Pezzolla et al., 1991; Riezzo et al., 1992; DiBaise et al.,
it is a clear that a rise in plasma vasopressin is not essential 2001). Few studies have been conducted on gastric
for the production of nausea under all circumstances and dysrhythmia in animal models of malaise. Gastric dysrhyth-
taken together with the studies of vasopressin infusion and mia precedes vomiting in the dog induced by electrical
plasma levels could indicate that vasopressin requires an stimulation of the vagus (Ueno and Chen, 2004), and
additional factor(s) to be present for the induction of nausea intravenous infusion of vasopressin produces gastric dys-
when it is released at a lower concentration. This appears to rhythmia in the dog (Xu et al., 2005).
108 P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

It is unknown whether gastric dysrhythmia, or neuro- 4. Neural circuitry for nausea


hypophyseal hormonal release, has any causative role in
the genesis of nausea or malaise produced by a broad Much is known about the neural system responsible for
range of stimuli. Furthermore, although the acute associ- emesis. Input systems that activate the vomiting reflex
ation between gastric dysrhythmia, or neurohypophyseal include cerebral, vestibular, area postrema (also known as
secretions, and nausea is well established more research the chemoreceptor trigger zone, although this name should
needs to focus on whether these physiological parameters not be taken to mean that all systemic agents inducing
are involved in delayed or chronic nausea and malaise. emesis act here), cardiac, and gastrointestinal vagal
Reports indicate that gastric dysrhythmia is associated with afferent fibres. These input systems activate a distributed
unexplained chronic nausea and vomiting (You et al., brainstem system, known collectively as the ‘‘vomiting
1981; Abell et al., 1988). centre.’’ The somatic and autonomic outputs of this system
are well established but the circuitry of the vomiting
centre is still a focus of intense research. In contrast to the
3. The role of pain in nausea and vomiting system for emesis relatively little is known about the
neural system responsible for nausea. This lack of
The perceptions of visceral pain and nausea are largely information is largely due to the difficulty in establishing
distinct. Visceral pain is a poorly localized phenomenon an animal model of nausea, as well as technical problems
that can be very intense. In contrast, nausea is an in studying this phenomenon in humans using brain
unsettling feeling or sensation of queasiness in the imaging techniques.
stomach. Although these phenomena appear to be separate, Although little is known about the neural system for
nausea is often associated with pain, and conversely pain nausea, this system appears to be anatomically distinct from
can produce nausea and vomiting. For example, postoper- the neural circuits responsible for emesis. It is clear that the
ative pain and nausea occur together, and when pain is vomiting reflex is contained within the brainstem because
reduced, nausea is also inhibited (Andersen and Krohg, even decerebrate animals are capable of vomiting and
1976). It is also not clear to what extent CFA (or learned emesis-like behaviour, e.g., the cat (Miller et al., 1994). It is
flavour aversion) or pica in laboratory animals measures likely that many of the autonomic responses that occur pre-
responses to pain, generalized stress, or a state of visceral and post-emesis are responses of the Fvomiting motor
malaise that might relate to the human perception of program_ or are responses to the Fstress_ of nausea. In fact
nausea. many of the autonomic signs of motion sickness can be
It is possible that nausea and visceral pain share some observed in a decorticate person on an airplane (Borison and
neural pathways or that one might modulate the other. The MacCarthy, 1983), and are unlikely to contribute to the
spinal nerves that innervate the gut play an important role genesis of nausea. Forebrain areas are required for learning
in producing visceral pain, and when these pathways are a flavour aversion in rat because decerebrate animals fail to
electrically stimulated humans report pain (White, 1943), show a conditioned response (Grill and Norgren, 1978).
whereas electrical stimulation of the vagal afferent fibres in However, although specific ablation of forebrain regions
humans can induce nausea (Lewis, 1942). Assuming that block CFA in the rat it is difficult to determine whether these
gut viscerosensory pathways are a necessary component for brain regions are important for the experience of malaise,
the genesis of nausea, spinal pathways might be involved and perhaps nausea in humans, or are involved in memory
in the production of nausea because nausea can persist in and learning processes of CFA (for review see Yamamoto et
humans even after sub-diaphragmatic vagotomy (Troncon al., 1994). Currently there are no published reports on the
et al., 1995). In contrast, the integrity of the vagal effects of brain or peripheral nerve lesions on pica.
pathways is necessary for the production of emesis by Nausea and emesis also appear to be pharmacologically
stimuli acting on the gut (Andrews et al., 1990). An separable. Despite the introduction of 5-HT3 receptor
alternative explanation for the persistence of nausea in antagonists (e.g. ondansetron, granisetron) for anti-emetic
patients with sub-diaphragmatic vagotomy is that stimula- therapy more than half of patients who receive chemo-
tion of cardiac vagal afferent fibres contributes to nausea in therapy continue to experience nausea (Herrington et al.,
these patients. Evidence suggests that stimulation of 2000; Morrow et al., 2000). Even with the newer,
cardiac vagal afferent fibres can evoke vomiting and tachykinin NK1 receptor antagonists (e.g., aprepitant) there
nausea (Abrahamsson and Thoren, 1973). Additionally, is less than complete control of the nausea component of
the nausea in vagotomized patients could have an chemotherapy (Andrews and Rudd, 2004). Nausea is often
endocrine origin (possibly via an action on the area considered more troublesome for patients because it can be
postrema) as nausea is associated with an increase in the present continuously in contrast to emesis which usually
plasma levels of a number of hormones including occurs in discrete episodes. It is usually assumed that
adrenaline (see above). It is also clear that pain can nausea is a low level stimulus that if increased would
enhance nausea, e.g., migraine headache or facial pain result in emesis. However, counter-intuitively, nausea is
increases nausea (Drummond and Granston, 2004, 2005). more difficult to block pharmacologically than emesis
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 109

(Morrow et al., 2002b), and nausea and vomiting can be have been most thoroughly investigated using cancer
dissociated experimentally by adjusting the stimulus chemotherapy agents (for review see Rudd and Andrews,
intensity. Nausea and vomiting also occur separately in a 2005). Many chemotherapy agents, such as cisplatin, cause
number of clinical situations; for example, raised intracra- the release of 5-HT from entero-endocrine cells (entero-
nial pressure induces emesis but is said to not be preceded chromaffin) of the gastrointestinal tract that leads to the
by nausea (Lee and Feldman, 1993) and some cancer activation of vagal afferent fibres containing 5-HT3 recep-
patients receiving radio-therapy and pregnant women may tors (Andrews et al., 1988; Minami et al., 1997; Hillsley and
experience vomiting without nausea (Tierson et al., 1986; Grundy, 1999). 5-HT3 receptor antagonists are very
Miralbell et al., 1995). In animal studies LiCl and CCK do effective blockers of the acute phase of vomiting and
not always produce emesis even at high doses when partially control the nausea produced by chemotherapy (e.g.,
malaise is suggested by behavioural (CFA) or physiolog- Cassidy et al., 1988; Massidda and Ionta, 1996; Morrow et
ical measures (elevated plasma vasopressin or oxytocin) al., 2002b). However, despite the introduction of 5-HT3
(Rabin and Hunt, 1992; Billig et al., 2001; Smith et al., receptor antagonists for anti-emetic therapy more than half
2001a). of patients who receive chemotherapy continue to experi-
Collectively, these results indicate that nausea and ence nausea (Herrington et al., 2000; Morrow et al., 2000).
vomiting are generated by neural systems that are at least Unlike emesis, 5-HT3 receptor antagonists might have their
partially separate. The brainstem is essential for the primary effect on nausea through direct action on the brain.
integration of the emetic signal and coordination of the These antagonist compounds readily cross the blood brain
motor components of emesis, and the projection of barrier where they could act on 5-HT3 receptors located
information rostrally from the brainstem to ‘‘higher’’ throughout the neuroaxis, although present in particularly
centres is required for the genesis of the sensation for high levels in the NTS.
nausea. Identification of the site at which the nausea and Abdominal vagal afferent fibres are not essential for the
emesis signals diverge in the brainstem is essential for sensation of nausea because even humans with bilateral
targeting therapeutic interventions likely to influence both abdominal vagotomy experience nausea, which might
nausea and vomiting. Whilst a common pathway for nausea suggest that spinal pathways play a role in the genesis of
and emesis might apply for stimuli acting via the vagal nausea by stimuli acting on the gut. There is little
afferent fibres and the area postrema, it is less clear that information on the role of visceral nerve pathways on
nausea requires the same neural routing via the brainstem animal studies of CFA or pica. Early reports showed that
compared to emesis originating from cerebral or vestibular abdominal vagotomy did not affect LiCl-induced CFA
sites. In this case, nausea originating from cerebral or (Martin et al., 1978), but did attenuate intra-gastric copper
vestibular sites might directly access the ‘‘higher’’ centres sulphate and motion-induced CFA in the rat (Coil et al.,
involved in the genesis of nausea without engaging the 1978; Fox and McKenna, 1988). There is no information on
brainstem circuits responsible for emesis (Rudd and the effects of vagal lesions on the acquisition of conditioned
Andrews, 2005). It is worth recalling that Dr. Kyrlov, a flavour aversion, using intra-oral infusion testing. Further-
colleague of Pavlov’s, described studies in the dog showing more, as mentioned above, there are no reports on the
that nausea, secretion of saliva, vomiting and sleep could effects of vagal lesions on toxicosis-induced pica.
all be induced by ‘‘the preliminaries of injection’’ following
injection of morphine itself over 5 or 6 days (Pavlov, 4.2. Direct action of stimuli on the brain?
1927). In some cases all the symptoms could be induced by
‘‘seeing the experimenter’’, an observation reminiscent of It is also possible that toxins and other treatments act on
the reports of nausea and vomiting induced in patients the brain directly to produce nausea. Activation of the area
when they have encountered the person who administered postrema has been suggested to play a role in CFA produced
chemotherapy outside a hospital setting. by copper sulphate and other blood borne toxins (Coil and
Norgren, 1981; Borison et al., 1984; Rabin et al., 1985).
4.1. Visceral afferent fibres However, evidence to support this hypothesis has relied on
AP ablation experiments, which, as noted above, are difficult
Vagal afferent neurons, connecting the gastrointestinal to interpret. These studies also did not directly measure
tract to the brain, play a large role in the generation of learned flavour aversion using the intra-oral infusion method.
vomiting (Andrews et al., 1990), and also serve as a Recent research showed that nausea associated with
pathway for the induction of nausea. In a biological as chemotherapy is correlated with a reduction of plasma
opposed to a clinical context, the upper gastrointestinal tract cortisol levels in cancer patients, which indicates an effect
vagal afferents are arguably most important post-ingestive (direct or indirect) on the hypothalamic-pituitary-adrenal axis
system for the induction of nausea (accompanied by reduced (Hursti et al., 1993; Morrow et al., 2002a). Studies in rat show
food intake, appetite and delayed gastric emptying) and elevated plasma cortisol levels with the production of CFA
vomiting if the stimulus is sufficiently intense. These (Smotherman, 1985). Additionally (as mentioned above),
peripheral nerve pathways for the initiation of vomiting there are many potential candidate hormones that might
110 P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

contribute to nausea, including vasopressin, oxytocin, adren- studied in species lacking an emetic reflex, that can be used to
aline, growth hormone, prolactin, adrenocorticotrophic hor- address this issue, however, there are likely to be significant
mone, and pancreatic polypeptide (Eversmann et al., 1978; overlaps between systems that generate emesis, CFA, and
Feldman et al., 1988; Nussey et al., 1988; Page et al., 1990; pica (or associated measures of plasma hormones and gastric
Xu et al., 1993). However, the challenge with all the systemic dysrythmia) with no guarantee that any of these responses
signals for nausea is to understand where and how they gain provide insights into the physiology of human nausea. It is
access to the central nervous system (e.g. via circumven- therefore important that results from animal experiments
tricular organs) and how they then induce the sensation of concerning nausea be validated in human studies and that the
nausea. It is also possible that some of these systemic agents results of such studies are in turn used to refine appropriate
act either on the gut to disrupt function and it is this which is models and abandon inappropriate ones. Ideally we should
signalled via the visceral afferents or that they act directly on work towards one widely accepted model of nausea and
receptors on the visceral afferent terminals. vomiting for pre-clinical testing of candidate therapies whilst
simultaneously increasing human studies particularly in the
4.3. The stimulus and coding of signals for nausea and area of nausea.
vomiting The current understanding of nausea and vomiting is also
limited by other factors that might be independent of species
Despite the well established role of the abdominal vagal selection. It is not clear how pain and stress are related to
afferents in emesis, and to a much lesser extent nausea, we nausea and vomiting. There are a large number of autonomic
have no clear understanding of the information code that is efferent and hormonal responses that accompany vomiting
conveyed to the brain by the visceral afferent fibres and how and the experience of nausea but many of these effects appear
this is encoded to signal nausea or induce vomiting. to be general Fstress_ responses. It also remains an important
Activation of the vagal afferent fibres by toxins in the gut question as to how the vagal afferent system codes
appears to operate via detection of toxins by enteroendo- information related to the genesis of nausea and vomiting.
crine cells that then release 5-HT as a paracrine factor that These unresolved issues guarantee that the study of nausea
activates 5-HT3 receptors on vagal afferent fibres. 5-HT and vomiting will remain an active area of research. This is
seems to be the predominant neurotransmitter in the particularly true because an understanding of the physiology
abdominal vagal system (Uneyama et al., 2002; Horn et and pharmacology of nausea and vomiting is of cardinal
al., 2004) although enteroendocrine cells such as the importance for developing Fclean_ drugs that target clinically
enterochromaffin cells which contain high levels of 5-HT important diseases as many potential therapeutic approaches
also contain substance P. There is neurophysiological have nausea and vomiting as dose limiting toxicities and also
evidence for complex interactions between 5-HT and SP for developing drugs acting to reduce both nausea and
and the activation of vagal afferent fibres via 5-HT3 and vomiting in a diverse range of clinical settings.
NK1 receptors (Minami et al., 2001). Evidence suggests that
serotonin signalling is also used for the detection of
nutrients, such as carbohydrates (Li et al., 2001; Zhu et Acknowledgements
al., 2001), which might serve to promote nutrient absorption
and processing and modify food intake. It is therefore The authors are supported by grants from the National
puzzling how the vagal system can accomplish these Institutes of Health (DK065971) and Glaxo Smith Kline.
multiple functions, including the sensing of toxins, using a
sensory system that appears to be highly likely to provide
ambiguous information to the brain.
References

Abell, T.L., Kim, C.H., Malagelada, J.R., 1988. Idiopathic cyclic nausea
5. Conclusions and vomiting—a disorder of gastrointestinal motility? Mayo Clin. Proc.
63, 1169 – 1175.
It is clear from the current review that investigating the Abrahamsson, H., Thoren, P., 1973. Vomiting and reflex vagal relaxation of
physiology of nausea and vomiting is very challenging, and the stomach elicited from heart receptors in the cat. Acta Physiol.
Scand. 88, 433 – 439.
much of this difficulty stems from limitations that exist when
Ader, R., 1976. Conditioned adrenocortical steroid elevations in the rat.
studying animal models. It is not known which species is J. Comp. Physiol. Psychol. 90, 1156 – 1163.
most predictive of the neuropharmacology of emesis in Amber, E.I., Henderson, R.A., Adeyanju, J.B., Gyang, E.O., 1990. Single-
humans when all types of emetic stimuli are considered, drug chemotherapy of canine transmissible venereal tumor with
although in the case of chemotherapy-induced emesis the cyclophosphamide, methotrexate, or vincristine. J. Vet. Intern. Med. 4,
ferret is probably the species used first to test a candidate anti- 144 – 147.
Andersen, R., Krohg, K., 1976. Pain as a major cause of postoperative
emetic agent. It is particularly difficult to decide what animal nausea. Can. Anaesth. Soc. J. 23, 366 – 369.
model is most appropriate for studying the symptom of Andrew, B.L., 1956. The nervous control of the cervical oesophagus of the
nausea. CFA and pica are potential model responses, usually rat during swallowing. J. Physiol. 134, 729 – 740.
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 111

Andrews, P.L.R., 1995. Why do some animals lack a vomiting reflex? Carr, D.H., Scott, P.C., Titchen, D.A., 1983. Manometric and
Physiol. Zool. 68, 61. electromyographic observations of the oesophagus of sheep in
Andrews, P.L.R., Rudd, J.A., 2004. The role of Tachykinins and the eructation, regurgitation and swallowing. Q. J. Exp. Physiol. 68,
Tachykinin NK1 receptor in nausea and emesis. In: Holzer, P. (Ed.), 661 – 674.
Tachykinins, Springer, Handbook of Experimental Pharmacology, Carrell, L.E., Cannon, D.S., Best, M.R., Stone, M.J., 1986. Nausea
pp. 359 – 440. and radiation-induced taste aversions in cancer patients. Appetite 7,
Andrews, P.L.R., Rapeport, W.G., Sanger, G.J., 1988. Neuropharmacology 203 – 208.
of emesis induced by anti-cancer therapy. Trends Pharmacol. Sci. 9, Cassidy, J., Raina, V., Lewis, C., Adams, L., Soukop, M., Rapeport, W.G.,
334 – 341. Zussman, B.D., Rankin, E.M., Kaye, S.B., 1988. Pharmacokinetics and
Andrews, P.L., Davis, C.J., Bingham, S., Davidson, H.I., Hawthorn, J., anti-emetic efficacy of BRL 43694, a new selective 5-HT3 antagonist.
Maskell, L., 1990. The abdominal visceral innervation and the emetic Br. J. Cancer 58, 651 – 653.
reflex: pathways, pharmacology, and plasticity. Can. J. Physiol. Pharm. Christian, M.S., York, R.G., Hoberman, A.M., Diener, R.M., Fisher, L.C.,
68, 325 – 345. Gates, G.A., 2001. Biodisposition of dibromoacetic acid (DBA) and
Andrews, P.L., Axelsson, M., Franklin, C., Holmgren, S., 2000. The emetic bromodichloromethane (BDCM) administered to rats and rabbits in
reflex in a reptile (Crocodylus porosus). J. Exp. Biol. 203, 1625 – 1632. drinking water during range-finding reproduction and developmental
Andrews, P.L., Kovacs, M., Watson, J.W., 2001. The anti-emetic action toxicity studies. Int. J. Toxicol. 20, 239 – 253.
of the neurokinin(1) receptor antagonist CP-99,994 does not require Coil, J.D., Norgren, R., 1981. Taste aversions conditioned with intravenous
the presence of the area postrema in the dog. Neurosci. Lett. 314, copper sulphate: attenuation by ablation of the area postrema. Brain
102 – 104. Res. 212, 425 – 433.
Andrews, P.L.R., Hoyle, C.H.V., Ngoka, T., Smith, G.E., 2003. Differences Coil, J.D., Hankins, W.G., Jenden, D.J., Garcia, J., 1978. The attenuation of
in the vagal innervation of the oesophagus may explain the lack of a specific cue-to-consequence association by antiemetic agents. Psy-
emesis in rodents. Neurogastroenterol. Motil. 15, 25. chopharmacology (Berlin) 56, 21 – 25.
Arwas, S., Rolnick, A., Lubow, R.E., 1989. Conditioned taste aversion in Cooper, J.R., Mattsson, J.L., 1979. Control of radiation-induced emesis
humans using motion-induced sickness as the US. Behav. Res. Ther. 27, with promethazine, cimetidine, thiethylperazine, or naloxone. Am. J.
295 – 301. Vet. Res. 40, 1057 – 1061.
Aung, H.H., Mehendale, S.R., Xie, J.T., Moss, J., Yuan, C.S., 2004. Corbett, R.W., Ryan, C., Weinrich, S.P., 2003. Pica in pregnancy: does it
Methylnaltrexone prevents morphine-induced kaolin intake in the rat. affect pregnancy outcomes? MCN, Am. J. Matern. Child Nursing 28,
Life Sci. 74, 2685 – 2691. 183 – 189.
Bailey, P.L., Rhondeau, S., Schafer, P.G., Lu, J.K., Timmins, B.S., Foster, Corcoran, M.L., Fox, R.A., Daunton, N.G., 1990. The susceptibility of
W., Pace, N.L., Stanley, T.H., 1993. Dose – response pharmacology of rhesus monkeys to motion sickness. Aviat. Space Environ. Med. 61,
intrathecal morphine in human volunteers. Anesthesiology 79, 49 – 59. 807 – 809.
Barnes, N.M., Bunce, K.T., Naylor, R.J., Rudd, J.A., 1991. The actions of Cordts, R.E., Yochmowitz, M.G., Hardy, K.A., 1987. Evaluation of
fentanyl to inhibit drug-induced emesis. Neuropharmacology 30, domperidone as a modifier of gamma-radiation-induced emesis. Int. J.
1073 – 1083. Radiat. Oncol. Biol. Phys. 13, 1333 – 1337.
Bayley, T.M., Dye, L., Jones, S., DeBono, M., Hill, A.J., 2002. Food Costall, B., Domeney, A.M., Naylor, R.J., 1987. A model of nausea and
cravings and aversions during pregnancy: relationships with nausea and emesis in the common marmoset. Br. J. Pharmacol. 88, 375.
vomiting. Appetite 38, 45 – 51. Costello, D.J., Borison, H.L., 1977. Naloxone antagonizes narcotic self
Beleslin, D.B., Krstic, S.K., Stefanovic-Denic, K., Strbac, M., Micic, D., blockade of emesis in the cat. J. Pharmacol. Exp. Ther. 203, 222 – 230.
1981. Inhibition by morphine and morphine-like drugs of nicotine- Crampton, G.H., Daunton, N.G., 1983. Systemic naloxone increases the
induced emesis in cats. Brain Res. Bull. 6, 451 – 453. incidence of motion sickness in the cat. Pharmacol. Biochem. Behav.
Bergman, J., Glowa, J.R., 1986. Suppression of behaviour by food pellet- 19, 827 – 829.
lithium chloride pairings in squirrel monkeys. Pharmacol. Biochem. Darolova, A., 1991. Food composition in the eagle-owl (Bubo bubo L.,
Behav. 25, 973 – 978. 1758) in Small Carpathians (Czechoslovakia). Biologia 45, 831 – 840.
Bielavska, E., Kren, V., Musilova, A., Zidek, V., Pravenec, M., 2002. Davis, C.J., Harding, R.K., Leslie, R.A., Andrews, P.L.R., 1986. The
Genome scanning of the HXB/BXH sets of recombinant inbred strains organisation of vomiting as a protective reflex. In: Davis, C.J., Lake-
of the rat for quantitative trait loci associated with conditioned taste Bakaar, G.V., Grahame-Smith, D.G. (Eds.), Nausea and Vomiting,
aversion. Behav. Genet. 32, 51 – 56. Mechanisms and Treatment. Springer-Verlag, Berlin, pp. 65 – 75.
Billig, I., Yates, B.J., Rinaman, L., 2001. Plasma hormone levels and Dawkins, R., 2004. The Ancestor’s Tale, Rodents and Rabbitkind.
central c-Fos expression in ferrets after systemic administration of Weidenfield & Nicholson, London, UK, pp. 152 – 161.
cholecystokinin. Am. J. Physiol., Regul. Integr. Comp. Physiol. 281, de Wit, R., Herrstedt, J., Rapoport, B., Carides, A.D., Guoguang-Ma, J.,
R1243 – R1255. Elmer, M., Schmidt, C., Evans, J.K., Horgan, K.J., 2004. The oral
Borison, H.L., MacCarthy, L.E., 1983. Neuropharmacologic mechanisms of NK(1) antagonist, aprepitant, given with standard antiemetics pro-
emesis. In: Laszlo, J. (Ed.), Anitemetics and Cancer Chemotherapy. vides protection against nausea and vomiting over multiple cycles of
Williams and Wilkins, Baltimore, pp. 6 – 20. cisplatin-based chemotherapy a combined analysis of two rando-
Borison, H.L., Borison, R., McCarthy, L.E., 1984. Role of the mised, placebo-controlled phase III clinical trials. Eur. J. Cancer 40,
area postrema in vomiting and related functions. Fed. Proc. 43, 403 – 410.
2955 – 2958. DiBaise, J.K., Brand, R.E., Lyden, E., Tarantolo, S.R., Quigley, E.M., 2001.
Braveman, N.S., 1974. Poison-based avoidance learning with flavoured or Gastric myoelectrical activity and its relationship to the development of
colored water in guinea pigs. Learn. Motiv. 5, 182 – 194. nausea and vomiting after intensive chemotherapy and autologous stem
Brizzee, K.R., 1956. Effect of localized brain stem lesions and supra- cell transplantation. Am. J. Gastroenterol. 96, 2873 – 2881.
diaphragmatic vagotomy on X-irradiation emesis in the monkey. Am. J. Dibattista, D., 1988. Conditioned taste aversion produced by 2-deoxy-d-
Physiol. 187, 567 – 570. glucose in rats and hamsters. Physiol. Behav. 44, 189 – 192.
Brizzee, K.R., Neal, L.M., Williams, P.M., 1955. The chemoreceptor trigger Dobbins, E.G., Feldman, J.L., 1994. Brainstem network controlling
zone for emesis in the monkey. Am. J. Physiol. 180, 659 – 662. descending drive to phrenic motoneurons in rat. J. Comp. Neurol. 347,
Caras, S.D., Soykan, I., Beverly, V., Lin, Z., McCallum, R.W., 1997. The 64 – 86.
effect of intravenous vasopressin on gastric myoelectrical activity in Drummond, P.D., Granston, A., 2004. Facial pain increases nausea and head-
human subjects. Neurogastroenterol. Motil. 9, 151 – 156. ache during motion sickness in migraine sufferers. Brain 127, 526 – 534.
112 P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

Drummond, P.D., Granston, A., 2005. Painful stimulation of the temple Gylys, J.A., Doran, K.M., Buyniski, J.P., 1979. Antagonism of cisplatin
induces nausea, headache and extracranial vasodilation in migraine induced emesis in the dog. Res. Commun. Chem. Pathol. Pharmacol.
sufferers. Cephalalgia 25, 16 – 22. 23, 61 – 68.
Edwards, C.M., Carmichael, J., Baylis, P.H., Harris, A.L., 1989. Arginine Harrison, W.A., Lipe, W.A., Decker, W.J., 1972. Apomorphine-induced
vasopressin—a mediator of chemotherapy induced emesis? Br. J. emesis in the dog: comparison of routes of administration. J. Am. Vet.
Cancer 59, 467 – 470. Med. Assoc. 160, 85 – 86.
Engel, C., 2002. Wild Health: How Animals Keep Themselves Well and Hasegawa, S., Takeda, N., Morita, M., Horii, A., Koizuka, I., Kubo, T.,
What We Can Learn From Them. Houghton Mifflin, New York. Matsunaga, T., 1992. Vestibular, central and gastral triggering of
Ervin, G.N., Mosher, J.T., Birkemo, L.S., Johnson, M.F., 1995. Multiple, emesis. A study on individual susceptibility in rats. Acta Oto-Laryngol.
small doses of cholecystokinin octapeptide are more efficacious at 112, 927 – 931.
inducing taste aversion conditioning than single, large doses. Peptides Hawthorn, J., Andrews, P.L., Ang, V.T., Jenkins, J.S., 1988. Differential
16, 539 – 545. release of vasopressin and oxytocin in response to abdominal vagal
Etscorn, F., 1980. Sucrose aversions in mice as a result of injected nicotine afferent stimulation or apomorphine in the ferret. Brain Res. 438,
or passive tobacco smoke inhalation. Bull. Psychon. Soc. 15, 54 – 56. 193 – 198.
Etscorn, F., Moore, G.A., Hagen, L.S., Caton, T.M., Sanders, D.L., 1986. Herrington, J.D., Kwan, P., Young, R.R., Lagow, E., Lagrone, L., Riggs,
Saccharin aversions in hamsters as a result of nicotine injections. M.W., 2000. Randomized, multicenter comparison of oral granisetron
Pharmacol. Biochem. Behav. 24, 567 – 570. and oral ondansetron for emetogenic chemotherapy. Pharmacotherapy
Etscorn, F., Moore, G.A., Scott, E.P., Hagen, L.S., Caton, T.M., Sanders, 20, 1318 – 1323.
D.L., Divine, K.K., 1987. Conditioned saccharin aversions in rats as a Hesketh, P.J., Van, B.S., Aapro, M., Tattersall, F.D., Naylor, R.J.,
result of cutaneous nicotine or intraperitoneal nicotine administered in Hargreaves, R., Carides, A.D., Evans, J.K., Horgan, K.J., 2003.
divided doses. Pharmacol. Biochem. Behav. 28, 495 – 502. Differential involvement of neurotransmitters through the time course
Eversmann, T., Gottsmann, M., Uhlich, E., Ulbrecht, G., von, W.K., Scriba, of cisplatin-induced emesis as revealed by therapy with specific
P.C., 1978. Increased secretion of growth hormone, prolactin, antidiu- receptor antagonists. Eur. J. Cancer 39, 1074 – 1080.
retic hormone, and cortisol induced by the stress of motion sickness. Hikami, K., Hasegawa, Y., Matsuzawa, T., 1990. Social transmission of
Aviat. Space Environ. Res. 49, 53 – 57. food preferences in Japanese monkeys (Macaca fuscata) after mere
Feldman, M., Samson, W.K., O’Dorisio, T.M., 1988. Apomorphine- exposure or aversion training. J. Comp. Psychol. 104, 233 – 237.
induced nausea in humans: release of vasopressin and pancreatic Hillsley, K., Grundy, D., 1999. Plasticity in the mesenteric afferent
polypeptide. Gastroenterology 95, 721 – 726. response to cisplatin following vagotomy in the rat. J. Auton. Nerv.
Fetting, J.H., McCarthy, L.E., Borison, H.L., Colvin, M., 1982. Vomiting Syst. 76, 93 – 98.
induced by cyclophosphamide and phosphoramide mustard in cats. Hobbs, S.H., Clingerman, H., Elkins, R.L., 1976. Illness-induced taste
Cancer Treat. Rep. 66, 1625 – 1629. aversions in normal and bulbectomized hamsters. Physiol. Behav. 17,
Fisher, R.D., Rentschler, R.E., Nelson, J.C., Godfrey, T.E., Wilbur, D.W., 235 – 238.
1982. Elevation of plasma antidiuretic hormones (ADH) associated with Holloway, R.H., 2001. Systemic pharmacomodulation of transient lower
chemotherapy-induced emesis in man. Cancer Treat. Rep. 66, 25 – 29. esophageal sphincter relaxations. Am. J. Med. 111, 178S – 185S.
Flanagan, L.M., Verbalis, J.G., Stricker, E.M., 1988. Naloxone potentiation Horn, C.C., Richardson, E.J., Andrews, P.L., Friedman, M.I., 2004.
of effects of cholecystokinin and lithium chloride on oxytocin secretion, Differential effects on gastrointestinal and hepatic vagal afferent fibres in
gastric motility and feeding. Neuroendocrinology 48, 668 – 673. the rat by the anti-cancer agent cisplatin. Auton. Neurosci. 115, 74 – 81.
Fox, R.A., 1977. Poison aversion and sexual behaviour in the golden Houpt, T.A., Pittman, D.W., Barranco, J.M., Brooks, E.H., Smith, J.C.,
hamster. Psychol. Rep. 41, 993 – 994. 2003. Behavioural effects of high-strength static magnetic fields on rats.
Fox, R.A., McKenna, S., 1988. Conditioned taste aversion induced by J. Neurosci. 23, 1498 – 1505.
motion is prevented by selective vagotomy in the rat. Behav. Neural Huang, W., Sved, A.F., Stricker, E.M., 2000. Vasopressin and oxytocin
Biol. 50, 275 – 284. release evoked by NaCl loads are selectively blunted by area
Fox, R.A., Corcoran, M., Brizzee, K.R., 1990. Conditioned taste aversion postrema lesions. Am. J. Physiol., Regul. Integr. Comp. Physiol. 278,
and motion sickness in cats and squirrel monkeys. Can. J. Physiol. R732 – R740.
Pharm. 68, 269 – 278. Hursti, T.J., Fredrikson, M., Steineck, G., Borjeson, S., Furst, C.J.,
Fukui, H., Yamamoto, M., Sasaki, S., Sato, S., 1993. Involvement of 5-HT3 Peterson, C., 1993. Endogenous cortisol exerts antiemetic effect similar
receptors and vagal afferents in copper sulphate-and cisplatin-induced to that of exogenous corticosteroids. Br. J. Cancer 68, 112 – 114.
emesis in monkeys. Eur. J. Pharmacol. 249, 13 – 18. Hutchison Jr., S.L., 1973. Taste aversion in albino rats using centrifugal
Furukawa, T., Yamada, K., 1980. The alpha-naphthoxyacetic acid-elicited spin as an unconditioned stimulus. Psychol. Rep. 33, 467 – 470.
retching involves dopaminergic inhibition in mice. Pharmacol. Bio- Ikegaya, Y., Matsuki, N., 2002. Vasopressin induces emesis in Suncus
chem. Behav. 12, 735 – 738. murinus. Jpn. J. Pharmacol. 89, 324 – 326.
Garcia, J., Kimeldorf, D.J., Koelling, R.A., 1955. Conditoned aversion to Jackson, T.R., Smith, J.W., 1978. A comparison of two aversion treatment
saccharin resulting from exposure to gamma radiation. Science 122, methods for alcoholism. J. Stud. Alcohol 39, 187 – 191.
157 – 158. Kayashima, N., Hayama, T., 1975. Reproducibility of copper sulphate
Gardner, C.J., Armour, D.R., Beattie, D.T., Gale, J.D., Hawcock, emesis by oral administration in dogs. Nippon Yakurigaku Zasshi 71,
A.B., Kilpatrick, G.J., Twissell, D.J., Ward, P., 1996. GR205171: a 169 – 173.
novel antagonist with high affinity for the tachykinin NK1 Kayashima, N., Hyama, T., 1976. Reproducibility of emesis by orally
receptor, and potent broad-spectrum anti-emetic activity. Regul. administrated copper sulphate in cats. Nippon Yakurigaku Zasshi 72,
Pept. 65, 45 – 53. 287 – 291.
Gibbs, R.A., et al., 2004. Genome sequence of the Brown Norway rat yields Kim, M.S., Chey, W.D., Owyang, C., Hasler, W.L., 1997. Role of plasma
insights into mammalian evolution. Nature 428, 493 – 521. vasopressin as a mediator of nausea and gastric slow wave dysrhyth-
Grant, P.J., Hughes, J.R., Dean, H.G., Davies, J.A., Prentice, C.R., 1986. mias in motion sickness. Am. J. Physiol. 272, G853 – G862.
Vasopressin and catecholamine secretion during apomorphine-induced Kimeldorf, D.J., Garcia, J., Rubadeau, D.O., 1960. Radiation-induced
nausea mediate acute changes in haemostatic function in man. Clin. Sci. conditioned avoidance behaviour in rats, mice, and cats. Radiat. Res.
(Lond.) 71, 621 – 624. 12, 710 – 718.
Grill, H.J., Norgren, R., 1978. Chronically decerebrate rats demonstrate King, G.L., 1990. Animal models in the study of vomiting. Can. J. Physiol.
satiation but not bait shyness. Science 201, 267 – 269. Pharm. 68, 260 – 268.
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 113

Kinney, N.E., Wright, J.W., Harding, J.W., 1993. Motion-induced aversions McCarthy, L.E., Borison, H.L., 1984. Cisplatin-induced vomiting elimi-
during and after recovery from olfactory nerve section in mice. Physiol. nated by ablation of the area postrema in cats. Cancer Treat. Rep. 68,
Behav. 53, 631 – 633. 401 – 404.
Koch, K.L., Stern, R.M., Vasey, M.W., Seaton, J.F., Demers, L.M., Harrison, McCutcheon, B., Ballard, M., McCaffrey, R.J., 1992. Intraperitoneally
T.S., 1990a. Neuroendocrine and gastric myoelectrical responses to injected cholecystokinin-octapeptide activates pica in rats. Physiol.
illusory self-motion in humans. Am. J. Physiol. 258, E304 – E310. Behav. 51, 543 – 547.
Koch, K.L., Summy-Long, J., Bingaman, S., Sperry, N., Stern, R.M., Miaskiewicz, S.L., Stricker, E.M., Verbalis, J.G., 1989. Neurohypophyseal
1990b. Vasopressin and oxytocin responses to illusory self-motion and secretion in response to cholecystokinin but not meal-induced gastric
nausea in man. J. Clin. Endocrinol. Metab. 71, 1269 – 1275. distention in humans. J. Clin. Endocrinol. Metab. 68, 837 – 843.
Krane, R.V., Sinnamon, H.M., Thomas, G.J., 1976. Conditioned taste Miller, A.D., Nonaka, S., Jakus, J., 1994. Brain areas essential or non-
aversions and neophobia in rats with hippocampal lesions. Comp. essential for emesis. Brain Res. 647, 255 – 264.
Physiol. Psychol. 90, 680 – 693. Minami, M., Ogawa, T., Endo, T., Hamaue, N., Hirafuji, M., Yoshioka, M.,
Kushner, R.F., Gleason, B., Shanta-Retelny, V., 2004. Reemergence of pica Blower, P.R., Andrews, P.L., 1997. Cyclophosphamide increases
following gastric bypass surgery for obesity: a new presentation of an 5-hydroxytryptamine release from the isolated ileum of the rat. Res.
old problem. J. Am. Diet. Assoc. 104, 1393 – 1397. Commun. Mol. Pathol. Pharmacol. 97, 13 – 24.
Lee, M., Feldman, M., 1993. Nausea and vomiting. In: Sleisenger, M.H., Minami, M., et al., 2001. Effects of CP-99, 994, a tachykinin NK(1)
Fordtran, J.S. (Eds.), Gastrointestinal Diseas. W.B. Saunders Co, receptor antagonist, on abdominal afferent vagal activity in ferrets:
Philadelphia, USA, pp. 509 – 523. evidence for involvement of NK(1) and 5-HT(3) receptors. Eur. J.
Lefebvre, R.A., Willems, J.L., Bogaert, M.G., 1981. Gastric relaxation and Pharmacol. 428, 215 – 220.
vomiting by apomorphine, morphine and fentanyl in the conscious dog. Miralbell, R., et al., 1995. Nausea and vomiting in fractionated radiother-
Eur. J. Pharmacol. 69, 139 – 145. apy: a prospective on-demand trial of tropisetron rescue for non-
Levine, A.S., Sievert, C.E., Morley, J.E., Gosnell, B.A., Silvis, S.E., 1984. responders to metoclopramide. Eur. J. Cancer 31A, 1461 – 1464.
Peptidergic regulation of feeding in the dog (Canis familiaris). Peptides Mitchell, D., 1976. Experiments on neophobia in wild and laboratory rats: a
5, 675 – 679. reevaluation. J. Comp. Physiol. Psychol. 90, 190 – 197.
Lewis, T., 1942. Pain. Macmillan, New York, USA, p. 192. Mitchell, D., Wells, C., Hoch, N., Lind, K., Woods, S.C., Mitchell, L.K.,
Li, B.U., Fleisher, D.R., 1999. Cyclic vomiting syndrome: features to be 1976. Poison induced pica in rats. Physiol. Behav. 17, 691 – 697.
explained by a pathophysiologic model. Dig. Dis. Sci. 44, 13S – 18S. Mitchell, D., Krusemark, M.L., Hafner, D., 1977a. Pica: a species relevant
Li, Y., Wu, X.Y., Zhu, J.X., Owyang, C., 2001. Intestinal serotonin acts as behavioural assay of motion sickness in the rat. Physiol. Behav. 18,
paracrine substance to mediate pancreatic secretion stimulated by 125 – 130.
luminal factors. Am. J. Physiol.: Gastrointest. Liver Physiol. 281, Mitchell, D., Winter, W., Morisaki, C.M., 1977b. Conditioned taste
G916 – G923. aversions accompanied by geophagia: evidence for the occurrence of
Limebeer, C.L., Parker, L.A., 1999. Delta-9-tetrahydrocannabinol interferes ‘‘psychological’’ factors in the etiology of pica. Psychosom. Med. 39,
with the establishment and the expression of conditioned rejection 401 – 412.
reactions produced by cyclophosphamide: a rat model of nausea. Mokri, B., 2004. Spontaneous low cerebrospinal pressure/volume head-
NeuroReport 10, 3769 – 3772. aches. Curr. Neurol. Neurosci. Rep. 4, 117 – 124.
Limebeer, C.L., Parker, L.A., 2000. The antiemetic drug ondansetron Money, K.E., 1970. Motion sickness. Physiol. Rev. 50, 1 – 35.
interferes with lithium-induced conditioned rejection reactions, but not Morrow, G.R., Andrews, P.L., Hickok, J.T., Stern, R., 2000. Vagal changes
lithium-induced taste avoidance in rats. J. Exp. Psychol., Anim. Behav. following cancer chemotherapy: implications for the development of
Processes 26, 371 – 384. nausea. Psychophysiology 37, 378 – 384.
Liu, J., Kashimura, S., Hara, K., Zhang, G., 2001. Death following cupric Morrow, G.R., Hickok, J.T., Andrews, P.L., Stern, R.M., 2002a. Reduction
sulphate emesis. J. Toxicol., Clin. Toxicol. 39, 161 – 163. in serum cortisol after platinum based chemotherapy for cancer: a role
Liu, Y.-L., Malik, N., Sanger, G.J., Friedman, M.I., Andrews, P.L.R., 2005. for the HPA axis in treatment-related nausea? Psychophysiology 39,
Pica—A model of nausea? Species differences in response to cisplatin. 491 – 495.
Physiol. Behav. 85, 271 – 277. Morrow, G.R., Roscoe, J.A., Hickok, J.T., Andrews, P.R., Matteson, S.,
Lockwood, D.R., Kwon, B., Smith, J.C., Houpt, T.A., 2003. Behavioural 2002b. Nausea and emesis: evidence for a biobehavioral perspective.
effects of static high magnetic fields on unrestrained and restrained Support. Care Cancer 10, 96 – 105.
mice. Physiol. Behav. 78, 635 – 640. Nakayama, H., Yamakuni, H., Nakayama, A., Maeda, Y., Imazumi, K.,
Lopez, L.B., Ortega Soler, C.R., de Portela, M.L., 2004. Pica during Matsuo, M., Mutoh, S., 2004. Diphenidol has no actual broad
pregnancy: a frequently underestimated problem. Arch. Latinoam. Nutr. antiemetic activity in dogs and ferrets. J. Pharmacol. Sci. 96, 301 – 306.
54, 17 – 24. Noble, R.L., 1945. Observations on various types of motion causing
Martin, J.R., Cheng, F.Y., Novin, D., 1978. Acquisition of learned taste vomiting in animals. Can. J. Res. 23, 212 – 225.
aversion following bilateral subdiaphragmatic vagotomy in rats. Norgren, R., Smith, G.P., 1988. Central distribution of subdiaphragmatic
Physiol. Behav. 21, 13 – 17. vagal branches in the rat. J. Comp. Neurol. 273, 207 – 223.
Massidda, B., Ionta, M.T., 1996. Prevention of delayed emesis by a single Nowlis, G.H., Frank, M.E., Pfaffmann, C., 1980. Specificity of acquired
intravenous bolus dose of 5-HT3-receptor-antagonist in moderately aversions to taste qualities in hamsters and rats. J. Comp. Physiol.
emetogenic chemotherapy. J. Chemother. 8, 237 – 242. Psychol. 94, 932 – 942.
Matsuki, N., Ueno, S., Kaji, T., Ishihara, A., Wang, C.H., Saito, H., 1988. Nussey, S.S., Hawthorn, J., Page, S.R., Ang, V.T., Jenkins, J.S., 1988.
Emesis induced by cancer chemotherapeutic agents in the Suncus Responses of plasma oxytocin and arginine vasopressin to nausea
murinus: a new experimental model. Jpn. J. Pharmacol. 48, 303 – 306. induced by apomorphine and ipecacuanha. Clin. Endocrinol. (Oxford)
Mattes, R.D., 1994. Prevention of food aversions in cancer patients during 28, 297 – 304.
treatment. Nutr. Cancer 21, 13 – 24. Page, S.R., Ang, V.T., Jackson, R., White, A., Nussey, S.S., Jenkins, J.S.,
Mattes, R.D., Curran Jr., W.J., Powlis, W., Whittington, R., 1991. A 1990. The effect of oxytocin infusion on adenohypophyseal function in
descriptive study of learned food aversions in radiotherapy patients. man. Clin. Endocrinol. (Oxford) 32, 307 – 313.
Physiol. Behav. 50, 1103 – 1109. Parker, L.A., 1995. Rewarding drugs produce taste avoidance, but not taste
Matteson, S., Roscoe, J., Hickok, J., Morrow, G.R., 2002. The role of aversion. Neurosci. Biobehav. Rev. 19, 143 – 157.
behavioural conditioning in the development of nausea. J. Obstet. Parker, L.A., Kwiatkowska, M., Burton, P., Mechoulam, R., 2004.
Gynecol. 186, S239. Effect of cannabinoids on lithium-induced vomiting in the Suncus
114 P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115

murinus (house musk shrew). Psychopharmacology (Berlin) 171, Saeki, M., Sakai, M., Saito, R., Kubota, H., Ariumi, H., Takano, Y.,
156 – 161. Yamatodani, A., Kamiya, H., 2001. Effects of HSP-117, a novel
Paton, J.F., Li, Y.W., Deuchars, J., Kasparov, S., 2000. Properties of solitary tachykinin NK1-receptor antagonist, on cisplatin-induced pica as a
tract neurons receiving inputs from the sub-diaphragmatic vagus nerve. new evaluation of delayed emesis in rats. Jpn. J. Pharmacol. 86, 359 –
Neuroscience 95, 141 – 153. 362.
Pavlov, I.P., 1927. Conditioned reflexes—an investigation of the physio- Santucci, D., Corazzi, G., Francia, N., Antonelli, A., Aloe, L., Alleva, E.,
logical activity of the cerebral cortex. Translated and edited by G.V. 2000. Neurobehavioural effects of hypergravity conditions in the adult
Anrep. Dover Edition 1960, Dover Publications Inc. New York, USA, mouse. NeuroReport 11, 3353 – 3356.
p. 430. Santucci, D., Francia, N., Aloe, L., Alleva, E., 2002. Neurobehavioural
Penagini, R., Bianchi, P.A., 1997. Effect of morphine on gastroesophageal responses to hypergravity environment in the CD-1 mouse. J. Gravit.
reflux and transient lower esophageal sphincter relaxation. Gastroente- Physiol. 9, 39 – 40.
rology 113, 409 – 414. Sarr, A.B., Mayura, K., Kubena, L.F., Harvey, R.B., Phillips, T.D., 1995.
Perera, A.D., Verbalis, J.G., Mikuma, N., Majumdar, S.S., Plant, T.M., 1993. Effects of phyllosilicate clay on the metabolic profile of aflatoxin B1 in
Cholecystokinin stimulates gonadotropin-releasing hormone release in Fischer-344 rats. Toxicol. Lett. 75, 145 – 151.
the monkey (Macaca mulatta). Endocrinology 132, 1723 – 1728. Schofferman, J.A., 1976. A clinical comparison of syrup of ipecac and
Pezzolla, F., Riezzo, G., Maselli, M.A., Giorgio, I., 1991. Gastric electrical apomorphine use in adults. JACEP 5, 22 – 25.
dysrhythmia following cholecystectomy in humans. Digestion 49, Schwartz, M.D., Jacobsen, P.B., Bovbjerg, D.H., 1996. Role of nausea in
134 – 139. the development of aversions to a beverage paired with chemotherapy
Phillips, T.D., 1999. Dietary clay in the chemoprevention of aflatoxin- treatment in cancer patients. Physiol. Behav. 59, 659 – 663.
induced disease. Toxicol. Sci. 52, 118 – 126. Selve, N., Friderichs, E., Reimann, W., Reinartz, S., 1994. Absence of
Phillips, T.D., Sarr, A.B., Grant, P.G., 1995. Selective chemisorption emetic effects of morphine and loperamide in Suncus murinus. Eur. J.
and detoxification of aflatoxins by phyllosilicate clay. Nat. Toxins 3, Pharmacol. 256, 287 – 293.
204 – 213. Sims, D.W., Andrews, P.L., Young, J.Z., 2000. Stomach rinsing in rays.
Rabin, B.M., Hunt, W.A., 1992. Relationship between vomiting and taste Nature 404, 566.
aversion learning in the ferret: studies with ionizing radiation, lithium Singer, L.K., York, D.A., Berthoud, H.R., Bray, G.A., 1999. Conditioned
chloride, and amphetamine. Behav. Neural Biol. 58, 83 – 93. taste aversion produced by inhibitors of fatty acid oxidation in rats.
Rabin, B.M., Hunt, W.A., Lee, J., 1985. Intragastric copper sulphate Physiol. Behav. 68, 175 – 179.
produces a more reliable conditioned taste aversion in vagotomized rats Sipols, A.J., Brief, D.J., Ginter, K.L., Saghafi, S., Woods, S.C., 1992.
than in intact rats. Behav. Neural Biol. 44, 364 – 373. Neuropeptide Y paradoxically increases food intake yet causes
Rabin, B.M., Hunt, W.A., Bakarich, A.C., Chedester, A.L., Lee, J., 1986a. conditioned flavour aversions. Physiol. Behav. 51, 1257 – 1260.
Angiotensin II-induced taste aversion learning in cats and rats and the Smith Jr., J.C., Halsted, J.A., 1970. Clay ingestion (geophagia) as a source
role of the area postrema. Physiol. Behav. 36, 1173 – 1178. of zinc for rats. J. Nutr. 100, 973 – 980.
Rabin, B.M., Hunt, W.A., Chedester, A.L., Lee, J., 1986b. Role of the area Smith, W.L., Alphin, R.S., Jackson, C.B., Sancilio, L.F., 1989. The
postrema in radiation-induced taste aversion learning and emesis in cats. antiemetic profile of zacopride. J. Pharm. Pharmacol. 41, 101 – 105.
Physiol. Behav. 37, 815 – 818. Smith, J.E., Friedman, M.I., Andrews, P.L., 2001a. Conditioned food
Reid, R.M., 1992. Cultural and medical perspectives on geophagia. Med. aversion in Suncus murinus (house musk shrew)— a new model for the
Anthropol. 13, 337 – 351. study of nausea in a species with an emetic reflex. Physiol. Behav. 73,
Reynolds, R.D., Binder, H.J., Miller, M.B., Chang, W.W., Horan, S., 593 – 598.
1968. Pagophagia and iron deficiency anemia. Ann. Intern. Med. 69, Smith, J.E., Paton, J.F., Andrews, P.L., 2001b. Cardiorespiratory reflexes in
435 – 440. a working heart-brainstem preparation of the house musk shrew, Suncus
Riezzo, G., Pezzolla, F., Darconza, G., Giorgio, I., 1992. Gastric murinus. Auton. Neurosci. 89, 54 – 59.
myoelectrical activity in the first trimester of pregnancy: a cutaneous Smotherman, W.P., 1985. Glucocorticoid and other hormonal substrates of
electrogastrographic study. Am. J. Gastroenterol. 87, 702 – 707. conditioned taste aversion. Ann. N.Y. Acad. Sci. 443, 126 – 144.
Risinger, F.O., Brown, M.M., 1996. Genetic differences in nicotine-induced Spealman, R.D., 1983. Maintenance of behaviour by postponement of
conditioned taste aversion. Life Sci. 58, 223 – 229. scheduled injections of nicotine in squirrel monkeys. J. Pharmacol. Exp.
Risinger, F.O., Cunningham, C.L., 2000. DBA/2J mice develop stronger Ther. 227, 154 – 159.
lithium chloride-induced conditioned taste and place aversions than Stephan, F.K., Smith, J.C., Fisher, E., 1999. Profound conditioned taste
C57BL/6J mice. Pharmacol. Biochem. Behav. 67, 17 – 24. aversion induced by oral consumption of 2-deoxy-D-glucose. Physiol.
Root-Bernstein, R., Root-Bernstein, M., 2000. Honey, mud, and maggots, Behav. 68, 221 – 226.
and other medical marvels: Pan Books, Macmillan Publishers Ltd. Stern, R.M., Koch, K.L., Stewart, W.R., Lindblad, I.M., 1987. Spectral
UK. analysis of tachygastria recorded during motion sickness. Gastroenter-
Roselle, H.A., 1970. Association of laundry starch and clay ingestion with ology 92, 92 – 97.
anemia in New York City. Arch. Intern. Med. 125, 57 – 61. Takeda, N., Hasegawa, S., Morita, M., Matsunaga, T., 1993. Pica in rats is
Rowe, J.W., Shelton, R.L., Helderman, J.H., Vestal, R.E., Robertson, G.L., analogous to emesis: an animal model in emesis research. Pharmacol.
1979. Influence of the emetic reflex on vasopressin release in man. Biochem. Behav. 45, 817 – 821.
Kidney Int. 16, 729 – 735. Takeda, N., Hasegawa, S., Morita, M., Horii, A., Uno, A., Yamatodani, A.,
Rudd, J.A., Andrews, P.L.R., 2005. Mechanisms of acute, delayed,and Matsunaga, T., 1995. Neuropharmacological mechanisms of emesis. II.
anticipatory emesis induced by anticancer therapies. In: Hesketh, P.J. Effects of antiemetic drugs on cisplatin-induced pica in rats. Methods
(Ed.), Management of Nausea and Vomiting in Cancer and Cancer Find. Exp. Clin. Pharmacol. 17, 647 – 652.
Treatment. Jones and Bartlett, Sudbury, MA, pp. 15 – 65. Tanihata, S., Oda, S., Nakai, S., Uchiyama, T., 2004. Antiemetic effect of
Rudd, J.A., Ngan, M.P., Wai, M.K., 1998. 5-HT3 receptors are not involved dexamethasone on cisplatin-induced early and delayed emesis in the
in conditioned taste aversions induced by 5-hydroxytryptamine, pigeon. Eur. J. Pharmacol. 484, 311 – 321.
ipecacuanha or cisplatin. Eur. J. Pharmacol. 352, 143 – 149. Tierson, F.D., Olsen, C.L., Hook, E.B., 1986. Nausea and vomiting of
Rudd, J.A., Yamamoto, K., Yamatodani, A., Takeda, N., 2002. Differential pregnancy and association with pregnancy outcome. Am. J. Obstet.
action of ondansetron and dexamethasone to modify cisplatin-induced Gynecol. 155, 1017 – 1022.
acute and delayed kaolin consumption (‘‘pica’’) in rats. Eur. J. Tonini, M., De, G.R., De, P.F., 2004. Progress with novel pharmacological
Pharmacol. 454, 47 – 52. strategies for gastro-oesophageal reflux disease. Drugs 64, 347 – 361.
P.L.R. Andrews, C.C. Horn / Autonomic Neuroscience: Basic and Clinical 125 (2006) 100 – 115 115

Torii, Y., Shikita, M., Saito, H., Matsuki, N., 1993. X-irradiation- induced illness in squirrel monkeys. Otolaryngol.-Head Neck Surg. 97,
induced emesis in Suncus murinus. J. Radiat. Res. (Tokyo) 34, 433 – 440.
164 – 170. Woods, S.C., Figlewicz, D.P., Madden, L., Porte Jr., D., Sipols, A.J., Seeley,
Troncon, L.E., Thompson, D.G., Ahluwalia, N.K., Barlow, J., Heggie, L., R.J., 1998. NPY and food intake: discrepancies in the model. Regul.
1995. Relations between upper abdominal symptoms and gastric Pept. 75-76, 403 – 408.
distension abnormalities in dysmotility like functional dyspepsia and Wu, M., Harding, R.K., Hugenholtz, H., Kucharczyk, J., 1985. Emetic
after vagotomy. Gut 37, 17 – 22. effects of centrally administered angiotensin II, arginine vasopressin
Tsukada, H., Hirose, T., Yokoyama, A., Kurita, Y., 2001. Randomised and neurotensin in the dog. Peptides 6 (Suppl. 1), 173 – 175.
comparison of ondansetron plus dexamethasone with dexamethasone Xu, L.H., Koch, K.L., Summy-Long, J., Stern, R.M., Seaton, J.F., Harrison,
alone for the control of delayed cisplatin-induced emesis. Eur. J. Cancer T.S., Demers, L.M., Bingaman, S., 1993. Hypothalamic and gastric
37, 2398 – 2404. myoelectrical responses during vection-induced nausea in healthy
Ueno, T., Chen, J.D., 2004. Vomiting and gastric electrical dysrhythmia in Chinese subjects. Am. J. Physiol. 265, E578 – E584.
dogs. Scand. J. Gastroenterol. 39, 344 – 352. Xu, X., Brining, D.L., Chen, J.D., 2005. Effects of vasopressin and long
Ueno, S., Matsuki, N., Saito, H., 1987. Suncus murinus: a new pulse-low frequency gastric electrical stimulation on gastric emptying,
experimental model in emesis research. Life Sci. 41, 513 – 518. gastric and intestinal myoelectrical activity and symptoms in dogs.
Ueno, S., Matsuki, N., Saito, H., 1988. Suncus murinus as a new Neurogastroenterol. Motil. 17, 236 – 244.
experimental model for motion sickness. Life Sci. 43, 413 – 420. Yamamoto, T., Shimura, T., Sako, N., Yasoshima, Y., Sakai, N., 1994.
Uneyama, H., Niijima, A., Tanaka, T., Torii, K., 2002. Receptor subtype Neural substrates for conditioned taste aversion in the rat. Behav. Brain
specific activation of the rat gastric vagal afferent fibres to serotonin. Res. 65, 123 – 137.
Life Sci. 72, 415 – 423. Yamamoto, K., Matsunaga, S., Matsui, M., Takeda, N., Yamatodani, A.,
Uvnas-Moberg, K., 1998. Antistress pattern induced by oxytocin. News 2002a. Pica in mice as a new model for the study of emesis. Methods
Physiol. Sci. 13, 22 – 25. Find. Exp. Clin. Pharmacol. 24, 135 – 138.
Van Lawick-Goodall, H., Van Lawick-Goodall, J., 1970. Innocnet Killers. Yamamoto, K., Takeda, N., Yamatodani, A., 2002b. Establishment of an
Colloins, London, UK. animal model for radiation-induced vomiting in rats using pica. J.
Vavilova, N.M., Kassil, V.G., 1984. Conditioned reflex taste aversion during Radiat. Res. (Tokyo) 43, 135 – 141.
dog ontogeny. Zh. Vyssh. Nerv. Deiat. Im I.P. Pavlova 34, 662 – 668. Yamamoto, K., Ngan, M.P., Takeda, N., Yamatodani, A., Rudd, J.A.,
Vig, M.M., 1990. Nicotine poisoning in a dog. Vet. Hum. Toxicol. 32, 2004. Differential activity of drugs to induce emesis and pica
573 – 575. behaviour in Suncus murinus (house musk shrew) and rats. Physiol.
Villablanca, J.R., Harris, C.M., Burgess, J.W., de, A.I., 1984. Reassessing Behav. 83, 151 – 156.
morphine effects in cats: I. Specific behavioural responses in intact and Yates, B.J., Miller, A.D., Lucot, J.B., 1998. Physiological basis and
unilaterally brain-lesioned animals. Pharmacol. Biochem. Behav. 21, pharmacology of motion sickness: an update. Brain Res. Bull. 47,
913 – 921. 395 – 406.
Watson, P.J., Hawkins, C., McKinney, J., Beatey, S., Bartles, R.R., Rhea, Yates, B.J., Smail, J.A., Stocker, S.D., Card, J.P., 1999. Transneuronal
K., 1987. Inhibited drinking and pica in rats following 2-deoxy-D- tracing of neural pathways controlling activity of diaphragm motoneur-
glucose. Physiol. Behav. 39, 745 – 752. ons in the ferret. Neuroscience 90, 1501 – 1513.
Weigel, M.M., Weigel, R.M., 1989. Nausea and vomiting of early You, C.H., Chey, W.Y., Lee, K.Y., Menguy, R., Bortoff, A., 1981.
pregnancy and pregnancy outcome. An epidemiological study. Br. J. Gastric and small intestinal myoelectric dysrhythmia associated
Obstet. Gynaecol. 96, 1304 – 1311. with chronic intractable nausea and vomiting. Ann. Intern. Med. 95,
White, J., 1943. Sensory innervation of the viscera. Res. Publ. Assoc. 23, 449 – 451.
373 – 390. Zhu, J.X., Wu, X.Y., Owyang, C., Li, Y., 2001. Intestinal serotonin acts as a
Wilpizeski, C.R., Lowry, L.D., Green, S.J., Smith Jr., B.D., Melnick, H., paracrine substance to mediate vagal signal transmission evoked by
1987. Subjective concomitants of motion sickness: quantifying rotation- luminal factors in the rat. J. Physiol. 530, 431 – 442.

Das könnte Ihnen auch gefallen