ArticlePDF AvailableLiterature Review

Abstract and Figures

Serotonergic systems play an important and generalized role in regulation of sleep-wake states and behavioral arousal. Recent in vivo electrophysiologic recording studies in animals suggest that several different subtypes of serotonergic neurons with unique behavioral correlates exist within the brainstem raphe nuclei, raising the possibility that topographically organized subpopulations of serotonergic neurons may have unique behavioral or physiologic correlates and unique functional properties. We have shown that the stress-related and anxiogenic neuropeptide corticotropin-releasing factor can stimulate the in vitro neuronal firing rates of topographically organized subpopulations of serotonergic neurons within the dorsal raphe nucleus (DR). These findings are consistent with a wealth of behavioral studies suggesting that serotonergic systems within the DR are involved in the modulation of ongoing anxiety-related behavior and in behavioral sensitization, a process whereby anxiety- and fear-related behavioral responses are sensitized for a period of up to 24 to 48 h. The dorsomedial subdivision of the DR, particularly its middle and caudal aspects, has attracted considerable attention as a region that may play a critical role in the regulation of acute and chronic anxiety states. Future studies aimed at characterization of the molecular and cellular properties of topographically organized subpopulations of serotonergic neurons are likely to lead to major advances in our understanding of the role of serotonergic systems in stress-related physiology and behavior.
Content may be subject to copyright.
46
Ann. N.Y. Acad. Sci. 1018: 46–57 (2004). © 2004 New York Academy of Sciences.
doi: 10.1196/annals.1296.005
Anatomic and Functional Topography
of the Dorsal Raphe Nucleus
JOLANE K. ABRAMS, PHILIP L. JOHNSON, JACOB H. HOLLIS,
AND CHRISTOPHER A. LOWRY
University Research Centre for Neuroendocrinology, University of Bristol,
Bristol BS2 8HW, UK
A
BSTRACT
: Serotonergic systems play an important and generalized role in
regulation of sleep–wake states and behavioral arousal. Recent
in vivo
electro-
physiologic recording studies in animals suggest that several different subtypes
of serotonergic neurons with unique behavioral correlates exist within the
brainstem raphe nuclei, raising the possibility that topographically organized
subpopulations of serotonergic neurons may have unique behavioral or physi-
ologic correlates and unique functional properties. We have shown that the
stress-related and anxiogenic neuropeptide corticotropin-releasing factor can
stimulate the
in vitro
neuronal firing rates of topographically organized sub-
populations of serotonergic neurons within the dorsal raphe nucleus (DR).
These findings are consistent with a wealth of behavioral studies suggesting
that serotonergic systems within the DR are involved in the modulation of on-
going anxiety-related behavior and in behavioral sensitization, a process whereby
anxiety- and fear-related behavioral responses are sensitized for a period of up
to 24 to 48 h. The dorsomedial subdivision of the DR, particularly its middle
and caudal aspects, has attracted considerable attention as a region that may
play a critical role in the regulation of acute and chronic anxiety states. Future
studies aimed at characterization of the molecular and cellular properties of
topographically organized subpopulations of serotonergic neurons are likely to
lead to major advances in our understanding of the role of serotonergic systems
in stress-related physiology and behavior.
K
EYWORDS
: fear; anxiety; anatomy; stress; serotonin; 5-hydroxytryptamine;
5-HT; topographic
INTRODUCTION
Serotonergic systems within the brainstem raphe complex are topographically or-
ganized with respect to their anatomic and functional properties. This anatomic and
functional specificity raises the possibility that topographically organized subpopu-
lations of serotonergic neurons may have unique stress-related functional properties.
Although there are clear differences between the anatomic and functional properties
of the dorsal raphe nucleus (DR) and the median raphe nucleus (MnR),
1
in this re-
view we focus on the anatomic and functional topographic organization within the
Address for correspondence: Jolane K. Abrams, University Research Centre for Neuroendo-
crinology, University of Bristol, Marlborough Street, Bristol BS2 8HW, United Kingdom. Voice:
+
44 (0) 117-928-2181; fax:
+
44 (0) 117-928-2080.
e-mail: J.K.Abrams@bristol.ac.uk
47ABRAMS
et al.
: TOPOGRAPHY OF SEROTONERGIC SYSTEMS
DR, a brainstem raphe nucleus that contains an extensive group of serotonergic neu-
rons (estimated to number 235,000 in the human brain
2
) projecting in a topographi-
cally organized manner to forebrain and brainstem targets. Recent technical
advances have demonstrated that although the DR has been defined as a single nu-
cleus by cytologic criteria, it comprises multiple subdivisions that differ with respect
to the characteristics of individual serotonergic neurons found within them. Topo-
graphically organized subpopulations of serotonergic neurons within the DR differ
in their morphologic features, membrane and cellular properties, afferent regulation,
and efferent projection patterns. In this review, we evaluate whether or not these dif-
ferences among subpopulations of serotonergic neurons have important implications
for neural mechanisms underlying serotonergic modulation of stress-related physi-
ology and behavior. In addition, we provide atlases of the rat and mouse DR and a
proposed nomenclature that may prove valuable for future studies exploring the fine
anatomic and functional topography of this nucleus.
SEROTONERGIC NEURONS IN DIFFERENT SUBDIVISIONS OF THE
DR HAVE UNIQUE MORPHOLOGIC AND CELLULAR FEATURES
Serotonergic neurons in the rat DR appear in topographically organized groups
of small round cells, medium-sized fusiform and bipolar cells, large fusiform cells,
and very large multipolar cells.
3
Similarly, serotonergic neurons in the human DR
appear in topographically organized groups of round, ovoid, fusiform, and multi-
polar cells.
2
Although we cannot predict functional properties of serotonergic cells
based on morphologic features, the fact that serotonergic neurons with different mor-
phologic features are topographically organized within different subdivisions of the
DR suggests a potential link between structure and function. Recently developed and
recently optimized technical approaches, such as whole-cell patch-clamp electro-
physiologic studies in visualized brain slices, should allow us to begin to investigate
structure–function relationships.
In addition to regional differences in the morphology of topographically organized
subpopulations of serotonergic neurons, immunohistochemical co-localization studies
have demonstrated that topographically organized subpopulations of serotonergic neu-
rons co-express different neurotransmitters, neuropeptides, or other proteins. For ex-
ample, recent studies have identified a subpopulation of serotonergic neurons within
the dorsomedial subdivision of the DR (~
8.00 mm bregma) that also expresses cor-
ticotropin-releasing factor (CRF).
4
This same study revealed, consistent with previous
studies,
5
that this region of the DR projects to the central nucleus of the amygdala
(CeA), a stress-related forebrain structure that is part of a distributed central autonomic
control system. These observations, along with the high expression of neurokinin-1
immunoreactivity on neuronal fibers within the dorsomedial DR, suggest that this sub-
division may play a role in serotonergic modulation of stress-related physiology and
behavior in rats and in stress-related neuropsychiatric disorders in humans.
EFFERENT PROJECTIONS ARISING FROM THE DR
ARE TOPOGRAPHICALLY ORGANIZED
The midbrain raphe nuclei, including the DR and the MnR, are the main sources
of serotonergic innervation of forebrain stress circuits.
1
The most consistent finding
48 ANNALS NEW YORK ACADEMY OF SCIENCES
in anatomic studies is that serotonergic neurons within the DR are organized topo-
graphically in the rostrocaudal plane. That is, serotonergic neurons located more ros-
trally project selectively to specific forebrain sites (e.g., the caudate putamen,
substantia nigra, and virtually all neocortical regions), whereas serotonergic neurons
located more caudally project to other forebrain sites (e.g., the septum, hippocam-
pus, and entorhinal cortex
3,6–8
). Serotonergic neurons that project to the caudate
putamen do not project to the hippocampus, and vice versa.
9
These simple anatomic
observations suggest that topographically organized subpopulations of serotonergic
neurons have unique functional properties associated with modulation of specific
forebrain systems.
SEROTONERGIC NEURONS GIVE RISE TO COLLATERAL
PROJECTIONS TO FUNCTIONALLY RELATED TARGETS
The use of multiple retrograde tracers in a single animal has led to the proposal
that not only are serotonergic neurons topographically organized within the mid-
brain and pontine raphe nuclei, but that individual serotonergic neurons give rise to
collateral (branched) projections to functionally related targets, such as those in-
volved in stress responses.
1
These collateral serotonergic projections could allow
temporally coordinated modulation of specific neural circuits, such as those impli-
cated in central autonomic control, anxiety, and conditioned fear, which include the
CeA and the paraventricular nucleus (PVN) of the hypothalamus.
1
Collateral projec-
tions from serotonergic neurons innervating the CeA and PVN are located primarily
in medial regions of the middle DR and in bilateral clusters of cells near the cerebral
aqueduct more caudally.
5,10
These double retrograde-tracing studies and others
10–13
provide clear evidence for an anatomic topography within the DR and suggest that
individual serotonergic neurons give rise to functionally related target structures,
providing an anatomic basis for serotonergic modulation of distributed functional
systems within the brain.
AFFERENT PROJECTIONS INNERVATING THE DR
ARE TOPOGRAPHICALLY ORGANIZED
Different subdivisions of the DR receive afferent input from different sources. Ret-
rograde and anterograde studies provide evidence for specific afferents to the ventral
and dorsal parts of the middle (~
8.00 mm bregma) DR, the rostral DR (~
7.30 mm
bregma), and the ventrolateral DR.
14,15
A recent immunohistochemical study in rats
found that CRF-ir fibers in the DR are organized along a rostral-to-caudal gradient and
that CRF-ir fibers innervating different subdivisions of the DR have different propor-
tions of excitatory versus inhibitory synaptic specializations, with the majority of syn-
apses between CRF-ir terminals and dendrites in the dorsolateral DR being excitatory
and the majority of those in the ventromedial region being inhibitory.
16
Although the
origin of the full complement of CRF-ir fibers innervating the DR is uncertain, a pro-
portion of CRF-ir afferents arises from the CeA, suggesting that they may play an im-
portant role in differential modulation of topographically organized subpopulations of
serotonergic neurons under stress-related conditions.
17
49ABRAMS
et al.
: TOPOGRAPHY OF SEROTONERGIC SYSTEMS
FUNCTIONAL TOPOGRAPHIC ORGANIZATION OF THE DR
Behavioral Correlates of Neuronal Activity
Some of the strongest evidence for topographic organization of subpopulations of
serotonergic neurons comes from studies combining electrophysiologic data with
behavioral observations. Single-unit extracellular recording studies in cats suggested
the existence of two different types of serotonergic neurons, designated type I and
type II serotonergic neurons, based on behavioral correlates. Type I serotonergic
neurons are active during waking arousal but show decreased activity during stages
of quiet waking (QW) and slow-wave sleep (SWS) and cease firing during paradoxic
sleep (PS) or rapid eye movement sleep. Type II serotonergic neurons are highly to-
pographically organized and are found only in small numbers in bilateral columns of
cells at the caudal interface of the dorsal and median raphe nuclei (reminiscent of the
interfascicular dorsal raphe nucleus in the rat brain). Type II serotonergic neurons,
like type I neurons, are active during waking arousal and QW but remain active dur-
ing SWS and PS.
18
Due to the overwhelming preponderance of type I serotonergic
neurons, it is likely that most conclusions drawn from single-unit electrophysiology
studies are drawn from random sampling of this population of cells.
A recent study sampling a large number of presumed serotonergic neurons within
the DR using single-unit recording methods has classified them into six different
subtypes based on their electroencephalographic and behavioral correlates in cats.
19
Serotonergic neurons were classified within two large categories: (1) typical seroto-
nergic neurons (e.g., similar to type I serotonergic neurons described previously) and
(2) atypical serotonergic neurons. Among the typical and atypical serotonergic neu-
rons, subpopulations were identified that may correspond to type I and type II sero-
tonergic neurons described previously (T
ABLE
1). According to Sakai
et al.
,
19
type
I-A and type I-B serotonergic neurons are evenly distributed in the DR, discharge
regularly at a high rate during active waking and at progressively slower rates during
SWS, and cease firing during SWS or PS (type I-A) or only during PS (type I-B);
atypical type I-C serotonergic neurons are unevenly distributed in the DR (located
predominantly within the ventral DR, similar to the atypical type II serotonergic neu-
rons) and discharge regularly at a high rate during active waking but have sustained
activity during SWS and PS. Other atypical serotonergic neurons (types II-A, II-B,
and II-C) have unique topographic distributions within the DR. Type II-A neurons
are located predominantly within the dorsomedial DR and show reductions in firing
during QW and SWS; unlike typical serotonergic neurons, the level of
α
1
-adrenergic
receptor activation does not seem to be a major determinant of neuronal activity during
SWS and PS. This study of a large sample of serotonergic neurons within the DR
points toward a greater degree of functional heterogeneity of serotonergic neurons
within the DR than had previously been suspected.
Cellular Properties and Afferent Control Mechanisms
Consistent with findings that the DR consists of multiple topographically orga-
nized subpopulations of serotonergic neurons with unique behavioral correlates,
studies investigating the responses of serotonergic neurons to specific afferent sig-
nals (neurotransmitters, neuropeptides) have demonstrated that responses are depen-
dent on the topographic location of the recordings.
50 ANNALS NEW YORK ACADEMY OF SCIENCES
TABLE 1. Typical and atypical serotonergic cells of the DR in freely moving cats (as described by Rasmussen
et al
.
18
and Sakai and
Crochet
19
)
Type Firing Pattern Distribution Behavioral Correlates % Cessation Blocked By
Typical groups
I-A (similar to Type I serotonergic
neurons in Rasmussen
et al
.
18
)
Clock-like Evenly distributed Cease firing during SWS with
PGO waves and PS
40 Histamine or
phenylephrine
I-B (similar to Type I serotonergic
neurons in Rasmussen
et al
.
18
)
Clock-like Evenly distributed Cease firing during SWS only 27 Histamine or
phenylephrine
Atypical groups
I-C (similar to Type II serotonergic
neurons in Rasmussen
et al
.
18
)
Clock-like Unevenly distributed
(ventral DR)
Sustained activity during SWS
and PS
6 Histamine or
phenylephrine
II-A Nonclock-like Unevenly distributed
(dorsomedial DR)
Reduction in firing during QW
and SWS. Ceased firing
before the occurrence of PGO
waves and remained silent
during SWS with PGO waves
and PS
8 Phenylephrine has no
effect; histamine
blocks cessation of
discharge during
sleep
II-B Nonclock-like Unevenly distributed
(rostral dorsal portion
of DR, near the
oculomotor n.)
Highest rate of discharge during
SWS with suppression of
activity during waking and PS;
strongly activated during
feeding or grooming
12 Phenylephrine
II-C Nonclock-like Unevenly distributed
(ventral DR near mlf
and nucleus annularis)
Sustained activity during PS 8 Histamine
A
BBREVIATIONS
: DR, dorsal raphe nucleus; SWS, slow-wave sleep; PGO, ponto-geniculo-occiptal; PS, paradoxic sleep; QW, quiet waking.
51ABRAMS
et al.
: TOPOGRAPHY OF SEROTONERGIC SYSTEMS
Electrophysiologic studies suggest that intracerebroventricular injection of CRF
at low doses inhibits the firing rates of DR 5-HT neurons
in vivo
in a topographically
dependent manner; the majority of the cells tested (60.7%) in the ventromedial DR
(
7.80 mm bregma) were inhibited by CRF, whereas responses of cells in the dorso-
medial and lateral wings (ventrolateral DR) were more variable.
20,21
In vitro
exper-
iments in our laboratory have found a greater proportion of CRF-responsive neurons
in the ventral midline of the caudal DR compared with the dorsal region of the DR
at the same rostrocaudal level (–8.00 to –8.40 mm bregma).
22
Recent studies using
visualized whole-cell patch-clamp electrophysiologic techniques reveal that sero-
tonergic neurons within the dorsomedial and ventromedial DR differ with respect
to active and passive membrane characteristics,
23
further supporting distinctions
among cellular properties and afferent control mechanisms of topographically orga-
nized serotonergic neurons within this nucleus.
Heterogeneity of 5-HT neuronal firing is also demonstrated in responses to com-
pounds related to circadian fluctuations in behavioral arousal. The cessation of
serotonergic neuronal firing in serotonergic neurons of the DR observed during PS
is not the result of GABA-mediated inhibition
24
but is rather the result of lack of
facilitation resulting from the local release of norepinephrine and histamine.
19
Dif-
ferent subtypes of serotonergic DR neurons respond differentially to microdialysis
application of phenylephrine (an
α
1
-adrenergic agonist) and histamine. Typical I-A
and I-B neurons’ normal quiescence during PS is blocked by phenylephrine or his-
tamine, whereas the atypical II-A and II-C neurons’ low firing rates are unaffected
by phenylephrine but are totally (II-A) or partially (II-C) blocked by histamine
(leading to substantial tonic activity during sleep).
19
These results strongly suggest
additional heterogeneity in the behavioral correlates and afferent control mecha-
nisms of topographically organized subpopulations of serotonergic neurons within
the DR.
Behavioral Studies
Anatomic and functional heterogeneity of the DR is nowhere more evident than
in the modulation of stress-related neural circuits and stress responses. Behavioral
studies involving site-specific injections of CRF into subregions of the DR support
the hypothesis that the DR has a functional topographic organization in the rostro-
caudal plane. A series of studies has demonstrated that increased serotonergic activity
arising from the DR plays a critical role in the development of learned helplessness,
including increases in behavioral measures of anxiety and fear measured 24 to 48 h
after exposure to inescapable stress.
25–27
Further studies have shown that these be-
havioral changes are dependent on CRF actions within the DR. Microinjection of the
nonselective CRF receptor antagonist D-Phe-CRF
(12–41)
into specific sites in the
caudal DR (particularly at ~–8.30 mm bregma) blocks behavioral changes induced
by inescapable stress.
28
Microinjections of CRF into the caudal DR (but not the ros-
tral DR) are sufficient to induce learned helplessness 24 h later in the absence of any
exposure to inescapable stress,
28
consistent with the hypothesis that CRF actions on
serotonergic systems arising from the caudal portion of the DR mediate these behav-
ioral changes. This is consistent with the finding that exposure to inescapable stress
increases the expression of the protein product of the immediate-early gene
c-fos
within the middle and caudal portions of the DR but not the rostral portion. This
52 ANNALS NEW YORK ACADEMY OF SCIENCES
FIGURE 1.
Anatomic atlas of tryptophan hydroxylase (TrpOH) immunohistochemical (ir)
staining in the rat DR. Sections (30
µ
m) were obtained from an adult male Wistar rat, and
every third section was immunostained with a sheep anti-TrpOH antibody. Bregma levels (mm)
are indicated in each photograph. Scale bar represents 50
µ
m. Anatomic levels assigned
based on comparisons with a rat stereotaxic atlas.
52
evidence points toward an essential role for the caudal DR in modulating the behav-
ioral consequences of inescapable stress.
IMMEDIATE-EARLY GENE EXPRESSION STUDIES
There is a large body of data concerning the effects of stressful stimuli on the ex-
pression of immediate-early genes within the DR.
29
Investigation of the effects of
stress-related stimuli on the expression of the protein products of immediate-early
genes such as
c-fos
is an effective means of evaluating the effects of a temporally
defined stimulus on large populations of cells, with resolution of functional responses
at the single-cell level. Paradigms associated with increased anxiety or conditioned
fear,
30–41
opiate withdrawal,
42,43
and intracerebroventricular infusion of CRF or
similar anxiogenic neuropeptides
44,45
are effective at increasing immediate-early
gene expression within caudal regions of the DR. When these data are taken to-
gether with those indicating increased serotonergic neurotransmission in the DR in
response to stressful stimuli,
46,47
especially when the stressor is intense, uncontrol-
lable, or unpredictable,
48–51
they suggest a mechanism whereby anxiogenic or fear-
inducing stimuli increase serotonergic neurotransmission via actions on subsets of
serotonergic neurons within the caudal regions of the DR that give rise to a meso-
limbocortical serotonergic innervation of the forebrain.
THE NEED FOR DEFINITIVE NEUROANATOMIC MAPS
With the goal of developing better treatments for human neuropsychiatric disor-
ders in mind, it is essential that rostrocaudal levels of the DR and other raphe nuclei
be carefully defined. A survey of the literature shows widely variable definitions,
with some groups’ definitions of “caudal” regions
20,21
being almost as far forward
as others’ definitions of “rostral” ones
22
. Therefore, we propose consensus defini-
tions based on standard stereotaxic atlases of the rat
52
and mouse
53
brain and our
own atlases of rat (F
IG
. 1) and mouse brain (F
IG
. 2) immunostaining with tryptophan
hydroxylase.
According to these atlases, the “rostral” DR (regions associated with the caudal
linear nucleus, ventrally) comprises anatomic levels from –6.92 to –7.64 mm bregma
in the rat brain (–4.12 to –4.48 mm bregma in the mouse); the “middle” DR includes
anatomic levels from –7.73 to –8.45 mm bregma in the rat brain (–4.54 to –4.90 mm
bregma in the mouse), and “caudal” DR includes anatomic levels from –8.54 to
–9.26 mm in the rat brain (–4.96 to –5.32 mm bregma in the mouse). We hope that
these maps will prove useful in making meaningful comparisons among different
studies of anatomic and functional subsets of serotonergic neurons in rodent brain.
53ABRAMS
et al.
: TOPOGRAPHY OF SEROTONERGIC SYSTEMS
54 ANNALS NEW YORK ACADEMY OF SCIENCES
FIGURE 2. Anatomic atlas of TrpOH-ir staining in the mouse DR. Sections (30 µm)
were obtained from an adult male Balb/c mouse, and every second section was immuno-
stained with a sheep anti-TrpOH antibody. Bregma levels (mm) are indicated in each photo-
graph. Scale bar represents 50 µm. Anatomic levels assigned based on comparisons with a
mouse stereotaxic atlas.53
55ABRAMS
et al.
: TOPOGRAPHY OF SEROTONERGIC SYSTEMS
ACKNOWLEDGMENTS
We gratefully acknowledge the support of the BBSRC (7/S14666), the NIMH
(R01MH065702-01A1), and the Neuroendocrinology Charitable Trust. C.A. Lowry
is a Wellcome Trust Research Fellow.
REFERENCES
1. L
OWRY
, C.A. 2002. Functional subsets of serotonergic neurones: implications for con-
trol of the hypothalamic-pituitary-adrenal axis J. Neuroendocrinol.
14:
911–923.
2. B
AKER
, K.G.,
et al.
1990. Cytoarchitecture of the human dorsal raphe nucleus. J.
Comp. Neurol.
301:
147–161.
3. S
TEINBUSCH
, H.W. 1984. Serotonin-immunoreactive neurons and their projections in
the CNS.
In
Handbook of Chemical Neuroanatomy, Vol. 3. A. Bjorklund & T. Hok-
felt, Eds.: 68–125. Elsevier. New York.
4. C
OMMONS
, K.G.,
et al.
2003. A neurochemically distinct dorsal raphe-limbic circuit
with a potential role in affective disorders. Neuropsychopharmacology
28:
206–215.
5. P
ETROV
, T.,
et al.
1994. Chemically defined collateral projections from the pons to the
central nucleus of the amygdala and hypothalamic paraventricular nucleus in the rat.
Cell Tissue Res.
277:
289–295.
6. S
TEINBUSCH
, H.W.,
et al.
1981. The nucleus raphe dorsalis of the rat and its projection
upon the caudatoputamen: a combined cytoarchitectonic, immunohistochemical and
retrograde transport study. J. Physiol. (Paris)
77:
157–174.
7. K
OHLER
, C.,
et al.
1982. Identification of serotonin and non-serotonin-containing neu-
rons of the mid-brain raphe projecting to the entorhinal area and the hippocampal
formation: a combined immunohistochemical and fluorescent retrograde tracing
study in the rat brain. Neuroscience
7:
951–975.
8. VERTES, R.P. 1991. A PHA-L analysis of ascending projections of the dorsal raphe
nucleus in the rat. J. Comp. Neurol. 313: 643–668.
9. IMAI, H., et al. 1986. The organization of divergent axonal projections from the mid-
brain raphe nuclei in the rat. J. Comp. Neurol. 243: 363–380.
10. PETROV, T., et al. 1992. The hypothalamic paraventricular and lateral parabrachial
nuclei receive collaterals from raphe nucleus neurons: a combined double retrograde
and immunocytochemical study. J. Comp. Neurol. 318: 18–26.
11. VILLAR, M.J., et al. 1988. Dorsal raphe serotoninergic branching neurons projecting
both to the lateral geniculate body and superior colliculus: a combined retrograde
tracing-immunohistochemical study in the rat. J. Comp. Neurol. 277: 126–140.
12. WILSON, M.A., et al. 1991. The organization of serotonergic projections to cerebral
cortex in primates: retrograde transport studies Neuroscience 44: 555–570.
13. LI, Y.Q., et al. 1993. Identification of periaqueductal gray and dorsal raphe nucleus
neurons projecting to both the trigeminal sensory complex and forebrain structures: a
fluorescent retrograde double-labeling study in the rat. Brain Res. 623: 267–277.
14. PEYRON, C., et al. 1996. Lower brainstem catecholamine afferents to the rat dorsal
raphe nucleus. J. Comp. Neurol. 364: 402–413.
15. PEYRON, C., et al. 1998. Forebrain afferents to the rat dorsal raphe nucleus demon-
strated by retrograde and anterograde tracing methods. Neuroscience 82: 443–468.
16. VALENTINO, R.J., et al. 2001. Evidence for regional heterogeneity in corticotropin-
releasing factor interactions in the dorsal raphe nucleus. J. Comp. Neurol. 435: 450–
463.
17. SAKANAKA, M., et al. 1987. Corticotropin releasing factor-like immunoreactivity in the
rat brain as revealed by a modified cobalt-glucose oxidase-diaminobenzidine
method. J. Comp. Neurol. 260: 256–298.
18. RASMUSSEN, K., et al. 1984. Activity of serotonin-containing neurons in nucleus cen-
tralis superior of freely moving cats. Exp. Neurol. 83: 302–317.
19. SAKAI, K., et al. 2001. Differentiation of presumed serotonergic dorsal raphe neurons
in relation to behavior and wake-sleep states. Neuroscience 104: 1141–1155.
56 ANNALS NEW YORK ACADEMY OF SCIENCES
20. PRICE, M.L., et al. 1998. Effects of corticotropin-releasing factor on brain serotonergic
activity. Neuropsychopharmacology 18: 492–502.
21. KIRBY, L.G., et al. 2000. Effects of corticotropin-releasing factor on neuronal activity
in the serotonergic dorsal raphe nucleus. Neuropsychopharmacology 22: 148–162.
22. LOWRY, C.A., et al. 2000. Corticotropin-releasing factor increases in vitro firing rates
of serotonergic neurons in the rat dorsal raphe nucleus: evidence for activation of a
topographically organized mesolimbocortical serotonergic system. J. Neurosci. 20:
7728–7736.
23. BECK, S.G. 2002. Median and dorsal raphe cellular characteristics: similarities and dif-
ferences [abstract]. 2002 Abstract Viewer/Itinerary Planner. Society for Neuroscience,
Washington, D.C. 636.4.
24. SAKAI, K., et al. 2000. Serotonergic dorsal raphe neurons cease firing by disfacilitation
during paradoxical sleep. Neuroreport 11: 3237–3241.
25. MAIER, S.F., et al. 1993. The role of the amygdala and dorsal raphe nucleus in mediat-
ing the behavioral consequences of inescapable shock. Behav. Neurosci. 107: 377–
388.
26. MAIER, S.F., et al. 1994. Chlordiazepoxide microinjected into the region of the dorsal
raphe nucleus eliminates the interference with escape responding produced by ines-
capable shock whether administered before inescapable shock or escape testing.
Behav. Neurosci. 108: 121–130.
27. MAIER, S.F., et al. 1995. 8-OH-DPAT microinjected in the region of the dorsal raphe
nucleus blocks and reverses the enhancement of fear conditioning and interference with
escape produced by exposure to inescapable shock. Behav. Neurosci. 109: 404–412.
28. HAMMACK, S.E., et al. 2002. The role of corticotropin-releasing hormone in the dorsal
raphe nucleus in mediating the behavioral consequences of uncontrollable stress. J.
Neurosci. 22: 1020–1026.
29. SENBA, E., et al. 1997. Stress-induced expression of immediate early genes in the brain
and peripheral organs of the rat. Neurosci. Res. 29: 183–207.
30. PEZZONE, M.A., et al. 1993. Activation of brainstem catecholaminergic neurons by
conditioned and unconditioned aversive stimuli as revealed by c-Fos immunoreactiv-
ity. Brain Res. 608: 310–318.
31. SILVEIRA, M.C., et al. 1993. Induction of Fos immunoreactivity in the brain by expo-
sure to the elevated plus-maze. Behav. Brain Res. 56: 115–118.
32. BECK, C.H., et al. 1995. Conditioned fear-induced changes in behavior and in the
expression of the immediate early gene c-fos: with and without diazepam pretreat-
ment. J. Neurosci. 15: 709–720.
33. MATSUDA, S., et al. 1996. Persistent c-fos expression in the brains of mice with chronic
social stress. Neurosci. Res. 26: 157–170.
34. BECKETT, S.R., et al. 1997. Central c-fos expression following 20kHz/ultrasound
induced defence behaviour in the rat. Brain Res. Bull. 42: 421–426.
35. CAMPEAU, S., et al. 1997. Neuroendocrine and behavioral responses and brain pattern
of c-fos induction associated with audiogenic stress. J. Neuroendocrinol. 9: 577–588.
36. KOLLACK-WALKER, S., et al. 1997. Social stress in hamsters: defeat activates specific
neurocircuits within the brain. J. Neurosci. 17: 8842–8855.
37. MARTINEZ, M., et al. 1998. Adaptation in patterns of c-fos expression in the brain asso-
ciated with exposure to either single or repeated social stress in male rats. Eur. J.
Neurosci. 10: 20–33.
38. NIKULINA, E.M., et al. 1998. Behavioral sensitization to cocaine after a brief social
stress is accompanied by changes in fos expression in the murine brainstem. Brain
Res. 810: 200–210.
39. CHUNG, K.K., et al. 1999. Central serotonin depletion modulates the behavioural,
endocrine and physiological responses to repeated social stress and subsequent c-fos
expression in the brains of male rats. Neuroscience 92: 613–625.
40. GRAHN, R.E., et al. 1999. Activation of serotonin-immunoreactive cells in the dorsal
raphe nucleus in rats exposed to an uncontrollable stressor. Brain Res. 826: 35–43.
41. CHUNG, K.K., et al. 2000. c-fos expression, behavioural, endocrine and autonomic
responses to acute social stress in male rats after chronic restraint: modulation by
serotonin. Neuroscience 95: 453–463.
57ABRAMS et al.: TOPOGRAPHY OF SEROTONERGIC SYSTEMS
42. CHIENG, B., et al. 1995. Increased fos-like immunoreactivity in the periaqueductal gray
of anaesthetised rats during opiate withdrawal. Neurosci. Lett. 183: 79–82.
43. CHAHL, L.A., et al. 1996. Distribution of c-Fos in guinea-pig brain following morphine
withdrawal. Brain Res. 717: 127–134.
44. VAUGHAN, J., et al. 1995. Urocortin, a mammalian neuropeptide related to fish uro-
tensin I and to corticotropin-releasing factor. Nature 378: 287–292.
45. BITTENCOURT, J.C., et al. 2000. Do centrally administered neuropeptides access cog-
nate receptors? An analysis in the central corticotropin-releasing factor system. J.
Neurosci. 20: 1142–1156.
46. ADELL, A., et al. 1997. Comparative study in the rat of the actions of different types of
stress on the release of 5-HT in raphe nuclei and forebrain areas. Neuropharmacol-
ogy 36: 735–741.
47. MASWOOD, S., et al. 1998. Exposure to inescapable but not escapable shock increases
extracellular levels of 5-HT in the dorsal raphe nucleus of the rat. Brain Res. 783:
115–120.
48. ADELL, A., et al. 1988. Time course of changes in serotonin and noradrenaline in rat
brain after predictable or unpredictable shock. Brain Res. 459: 54–59.
49. INOUE, T., et al. 1994. Regional changes in dopamine and serotonin activation with var-
ious intensity of physical and psychological stress in the rat brain. Pharmacol. Bio-
chem. Behav. 49: 911–920.
50. AMAT, J., et al. 1998. Escapable and inescapable stress differentially alter extracellular
levels of 5-HT in the basolateral amygdala of the rat. Brain Res. 812: 113–120.
51. AMAT, J., et al. 1998. Escapable and inescapable stress differentially and selectively
alter extracellular levels of 5-HT in the ventral hippocampus and dorsal periaqueduc-
tal gray of the rat. Brain Res. 797: 12–22.
52. PAXINOS, G., et al. 1997. The Rat Brain in Stereotaxic Coordinates. Academic Press.
San Diego, CA.
53. PAXINOS, G., et al. 2001. The Mouse Brain in Stereotaxic Coordinates, 2nd Edition.
Academic Press. San Diego, CA.
... There are also dense serotonergic projections between the DRN and MRN [11]. The central nervous system therefore contains a small number of 5-HT neurons grouped together in nuclei, but these neurons release serotonin diffusely throughout the brain at chemical synapses or by volume transmission [4,12]. Serotonin can therefore exert a modulating action on a wide variety of neuronal circuits and therefore functions [13,14]. ...
Article
Full-text available
In recent years, particular attention has been paid to the serotonin 4 receptor, which is well expressed in the brain, but also peripherally in various organs. The cerebral distribution of this receptor is well conserved across species, with high densities in the basal ganglia, where they are expressed by GABAergic neurons. The 5-HT4 receptor is also present in the cerebral cortex, hippocampus, and amygdala, where they are carried by glutamatergic or cholinergic neurons. Outside the central nervous system, the 5-HT4 receptor is notably expressed in the gastrointestinal tract. The wide distribution of the 5-HT4 receptor undoubtedly contributes to its involvement in a plethora of functions. In addition, the modulation of this receptor influences the release of serotonin, but also the release of other neurotransmitters such as acetylcholine and dopamine. This is a considerable asset, as the modulation of the 5-HT4 receptor can therefore play a direct or indirect beneficial role in various disorders. One of the main advantages of this receptor is that it mediates a much faster antidepressant and anxiolytic action than classical selective serotonin reuptake inhibitors. Another major benefit of the 5-HT4 receptor is that its activation enhances cognitive performance, probably via the release of acetylcholine. The expression of the 5-HT4 receptor is also altered in various eating disorders, and its activation by the 5-HT4 agonist negatively regulates food intake. Additionally, although the cerebral expression of this receptor is modified in certain movement-related disorders, it is still yet to be determined whether this receptor plays a key role in their pathophysiology. Finally, there is no longer any need to demonstrate the value of 5-HT4 receptor agonists in the pharmacological management of gastrointestinal disorders.
... Supporting the regional (Fig. 3b) and AD subtype specificities ( Supplementary Fig. 10a) of spatial cluster 1, Glu signaling is predominantly seen in hippocampal regions and the surrounding medial temporal cortex [83], and Gluergic modulation has been implicated in OCD pathogenesis [84]. On the other hand, 5-HT production is confined to the midbrain raphe nuclei [85] and the ventral tegmental area is a hub for DA production [86], both midbrain areas associated with spatial cluster 2 (Fig. 3c). 5-HT and DA play important roles in AD subtypes affiliated with spatial cluster 2. In PD patients, 5-HT agonists induced anxiogenic responses, whereas selective 5-HT reuptake inhibitors showed therapeutic benefits [87]. ...
Article
Full-text available
Anxiety disorders (ADs) are the most common form of mental disorder that affects millions of individuals worldwide. Although physiological studies have revealed the neural circuits related to AD symptoms, how AD-associated genes are spatiotemporally expressed in the human brain still remains unclear. In this study, we integrated genome-wide association studies of four human AD subtypes—generalized anxiety disorder, social anxiety disorder, panic disorder, and obsessive-compulsive disorder—with spatial gene expression patterns. Our investigation uncovered a novel division among AD-associated genes, marked by significant and distinct expression enrichments in the cerebral nuclei, limbic, and midbrain regions. Each gene cluster was associated with specific anxiety-related behaviors, signaling pathways, region-specific gene networks, and cell types. Notably, we observed a significant negative correlation in the temporal expression patterns of these gene clusters during various developmental stages. Moreover, the specific brain regions enriched in each gene group aligned with neural circuits previously associated with negative decision-making and anxious temperament. These results suggest that the two distinct gene clusters may underlie separate neural systems involved in anxiety. As a result, our findings bridge the gap between genes and neural circuitry, shedding light on the mechanisms underlying AD-associated behaviors.
Article
Full-text available
Research highlights the significance of increased bursting in lateral habenula (LHb) neurons in depression and as a focal point for bright light treatment (BLT). However, the precise spike patterns of LHb neurons projecting to different brain regions during depression, their roles in depression development, and BLT's therapeutic action remain elusive. Here, LHb neurons are found projecting to the dorsal raphe nucleus (DRN), ventral tegmental area (VTA), and median raphe nucleus (MnR) exhibit increased bursting following aversive stimuli exposure, correlating with distinct depressive symptoms. Enhanced bursting in DRN‐projecting LHb neurons is pivotal for anhedonia and anxiety, while concurrent bursting in LHb neurons projecting to the DRN, VTA, and MnR is essential for despair. Remarkably, reducing bursting in distinct LHb neuron subpopulations underlies the therapeutic effects of BLT on specific depressive behaviors. These findings provide valuable insights into the mechanisms of depression and the antidepressant action of BLT.
Article
Full-text available
Obesity, associated with the intake of a high-fat diet (HFD), and anxiety are common among those living in modern urban societies. Recent studies suggest a role of microbiome-gut-brain axis signaling, including a role for brain serotonergic systems in the relationship between HFD and anxiety. Evidence suggests the gut microbiome and the serotonergic brain system together may play an important role in this response. Here we conducted a nine-week HFD protocol in male rats, followed by an analysis of the gut microbiome diversity and community composition, brainstem serotonergic gene expression (tph2, htr1a, and slc6a4), and anxiety-related defensive behavioral responses. We show that HFD intake decreased alpha diversity and altered the community composition of the gut microbiome in association with obesity, increased brainstem tph2, htr1a and slc6a4 mRNA expression, including in the caudal part of the dorsomedial dorsal raphe nucleus (cDRD), a subregion previously associated with stress- and anxiety-related behavioral responses, and, finally, increased anxiety-related defensive behavioral responses. The HFD increased the Firmicutes/Bacteroidetes ratio relative to control diet, as well as higher relative abundances of Blautia, and decreases in Prevotella. We found that tph2, htr1a and slc6a4 mRNA expression were increased in subregions of the dorsal raphe nucleus in the HFD, relative to control diet. Specific bacterial taxa were associated with increased serotonergic gene expression in the cDRD. Thus, we propose that HFD-induced obesity is associated with altered microbiome-gut-serotonergic brain axis signaling, leading to increased anxiety-related defensive behavioral responses in rats. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-024-00505-1.
Article
Dysfunction of the brain serotonergic system is thought to play a leading role in the pathogenesis of chronic abdominal pain and comorbid somatic hyperalgesia, which disturb a significant proportion of patients with digestive tract diseases, even in remission. However, the specific changes in the serotonergic structures nociceptive properties that can be initiated by organic pathology and persist after its resolution remain unclear. The aim of our neurophysiological study on anesthetized rats – healthy and recovered from colitis – was to identify the alterations in the raphe magnus (RMg) and dorsal raphe (DR) nuclei neuronal responses to visceral (colorectal distension) and somatic (squeezing of the tail) noxious stimulations that persist after resolution of intestinal inflammation. It has been shown that both nuclei contain different groups of nociceptive neurons: 1) responding with activation only to colorectal distension (visceral); 2) excited only by tail squeezing (somatic); 3) reacting with excitation to the both irritations (general); 4) responding with discharge inhibition to any of the stimulations (inhibited). Compared with healthy animals, in RMg of colitis-exposed rats the number of the inhibited cells was increased and the total proportion of excited nociceptive neurons was reduced. Distension of the inflammation-undergone intestine caused enhanced RMg neuronal inhibition, whereas squeezing of the pathology-unaffected tail led to increased excitation of the RMg selective somatic and general nociceptive cells. In turn, in the DR of postcolitis rats the inhibited neuron proportion was reduced, while the increased population of excited neurons included fewer visceral and more somatic selective cells. This was accompanied by an increase in the selective reactions of the latter to somatic pain stimuli and by an increase in non-selective DR neuron excitation by visceral and somatic pain signals. The identified neuronal alterations can contribute to the postcolitis impairment of the studied raphe nuclei functions in the endogenous control of visceral and somatic pain sensitivity.
Article
Anxiety disorders are the most common psychiatric condition, but the etiology of anxiety disorders remains largely unclear. Our previous studies have shown that neuroplastin 65 deficiency (NP65-/-) mice exhibit abnormal social and mental behaviors and decreased expression of tryptophan hydroxylase 2 (TPH2) protein. However, whether a causal relationship between TPH2 reduction and anxiety disorders exists needs to be determined. In present study, we found that replenishment of TPH2 in dorsal raphe nucleus (DRN) enhanced 5-HT level in the hippocampus and alleviated anxiety-like behaviors. In addition, injection of AAV-NP65 in DRN significantly increased TPH2 expression in DRN and hippocampus, and reduced anxiety-like behaviors. Acute administration of exogenous 5-HT or HTR3 agonist SR57227A in hippocampus mitigated anxiety-like behaviors in NP65-/- mice. Moreover, replenishment of TPH2 in DRN partly repaired the impairment of long-term potentiation (LTP) maintenance in hippocampus of NP65-/- mice. Finally, we found that loss of NP65 lowered transcription factors Lmx1b expression in postnatal stage and replenishment of NP65 in DRN reversed the decrease in Lmx1b expression of NP65-/- mice. Together, our findings reveal that NP65 deficiency induces anxiety phenotype by downregulating DRN-hippocampus serotonergic-HTR3 transmission. These studies provide a novel and insightful view about NP65 function, suggesting an attractive potential target for treatment of anxiety disorders.
Article
The anteroventral bed nucleus of the stria terminalis (avBNST) is widely acknowledged as a key brain structure that regulates negative emotional states, such as anxiety. At present, it is still unclear whether GABAA receptor-mediated inhibitory transmission in the avBNST is involved in Parkinson's disease (PD)-related anxiety. In this study, unilateral 6-hydroxydopamine (6-OHDA) lesions of the substantia nigra pars compacta (SNc) in rats induced anxiety-like behaviors, increased GABA synthesis and release, and upregulated expression of GABAA receptor subunits in the avBNST, as well as decreased level of dopamine (DA) in the basolateral amygdala (BLA). In both sham and 6-OHDA rats, intra-avBNST injection of GABAA receptor agonist muscimol induced the following changes: (i) anxiolytic-like responses, (ii) inhibition of the firing activity of GABAergic neurons in the avBNST, (iii) excitation of dopaminergic neurons in the ventral tegmental area (VTA) and serotonergic neurons in the dorsal raphe nucleus (DRN), and (iv) increase of DA and 5-HT release in the BLA, whereas antagonist bicuculline induced the opposite effects. Collectively, these findings suggest that degeneration of the nigrostriatal pathway enhances GABAA receptor-mediated inhibitory transmission in the avBNST, which is involved in PD-related anxiety. Further, activation and blockade of avBNST GABAA receptors affect the firing activity of VTA dopaminergic and DRN serotonergic neurons, and then change release of BLA DA and 5-HT, thereby regulating anxiety-like behaviors.
Article
Full-text available
Systemic administration of benzodiazepines before exposure to inescapable shock (IS) blocks the enhanced fear conditioning and escape learning deficits that follow exposure to IS, whereas administration before the subsequent behavioral testing eliminates the enhanced fear but not the interference with escape. The failure of benzodiazepines to reduce the IS-produced escape learning deficit when given before testing is inconsistent with a recent proposal that interference with escape is mediated by an IS-induced sensitization of dorsal raphe nucleus (DRN) activity. The present experiments demonstrate that chlordiazepoxide will block both the enhancement of fear and interference with escape responding when given before either IS or testing if microinjected in the region of the DRN. This suggests that systemic benzodiazepines fail to block escape deficits when given before testing because action at a site distant from the DRN counters the effect of the drug at the DRN.
Article
Full-text available
A large body of data suggests that the activation of α1 receptors by a tonic noradrenergic input might be responsible for the tonic discharge of the serotonergic neurons of the dorsal raphe nucleus (DRN). To test this hypothesis, it was necessary to determine the origin of the noradrenergic and adrenergic innervation of these neurons. For this purpose, we combined small iontophoretic injections of the sensitive retrograde tracer cholera toxin b subunit (CTb) in the different subdivisions of the DRN with tyrosine hydroxylase immunohistochemistry. After CTb injections in the ventral or dorsal parts of the central DRN, a small number of double-labeled cells was observed in the locus coeruleus (A6 noradrenergic cell group), the A5 noradrenergic group, the dorsomedial medulla (C3 adrenergic cell group), and the lateral paragigantocellular nucleus (C1 adrenergic cell group). After CTb injections in the lateral wings or the dorsal part of the rostral DRN, a similar number of double-labeled cells was seen in C3. Slightly more double-labeled cells were seen in A6 and A5. In addition, a substantial to large number of double-labeled cells appeared in C1, the commissural part of the nucleus of the solitary tract (A2 noradrenergic cell group) and the caudoventrolateral medulla (A1 noradrenergic cell group). These results indicate that the noradrenergic and adrenergic inputs to the DRN arise from all the catecholaminergic cell groups of the lower brainstem except the A7 noradrenergic group. They further reveal the existence of a topographical organization of these afferents to the different subdivisions of the DRN.
Article
Full-text available
CORTICOTROPIN-RELEASING factor (CRF), a peptide first isolated from mammalian brain1, is critical in the regulation of the pituitary–adrenal axis, and in complementary stress-related endocrine, autonomic and behavioural responses2. Fish urotensin I and amphibian sauvagine were considered to be homologues3 of CRF until peptides even more closely related to CRF were identified in these same vertebrate classes4,5. We have characterized another mammalian member of the CRF family and have localized its urotensin-like immunoreactivity to, and cloned related complementary DNAs from, a discrete rat midbrain region. The deduced protein encodes a peptide that we name urocortin, which is related to urotensin (63% sequence identity) and CRF (45% sequence identity). Synthetic urocortin evokes secretion of adrenocortico-tropic hormone (ACTH) both in vitro and in vivo and binds and activates transfected type-1 CRF receptors6–9, the subtype expressed by pituitary corticotropes. The coincidence of urotensin-like immunoreactivity with type-2 CRF receptors10–13 in brain, and our observation that urocortin is more potent than CRF at binding and activating type-2 CRF receptors, as well as at inducing c-Fos (an index of cellular activation) in regions enriched in type-2 CRF receptors, indicate that this new peptide could be an endogenous ligand for type-2 CRF receptors.
Article
Full-text available
It has been argued that exposure to inescapable shock produces later behavioral changes such as poor shuttle box escape learning because it leads to the conditioning of intense fear, which later transfers to the shuttle box test situation and interferes with escape. Both fear, as assessed by freezing, and escape were measured in Sprague-Dawley rats 24 hrs after exposure to inescapable shock. Lesions of the basolateral region and central nucleus of the amygdala eliminated the fear that transfers to the shuttle box after inescapable shock, as well as the fear conditioned in the shuttle box by the shuttle box shocks. However, the amygdala lesions did not reduce the learning deficit produced by inescapable shock. In contrast, dorsal raphe nucleus lesions did not reduce the fear that transfers to the shuttle box after inescapable shock, but eliminated the enhanced fear conditioning in the shuttle box as well as the escape deficit. The implications of these results for the role of fear and anxiety in mediating inescapable shock effects are discussed. (PsycINFO Database Record (c) 2012 APA, all rights reserved)