You are on page 1of 101

2000 Nature America Inc. http://neurosci.nature.

com

contents

volume 3 no 2 february 2000

http://neurosci.nature.com

Kim and colleagues report a tech-


nical advance that gives sufficient
spatial resolution to image cortical
editorial
column organization by fMRI in
2000 Nature America Inc. http://neurosci.nature.com

cats. Instead of the traditional How experts communicate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97


positive BOLD fMRI signal, the
authors used an earlier, negative
phase of the signal to obtain the
map of cortical orientation
columns shown (image filtered
news and views
and rearranged for artistic
purposes). See pages 105 and 164. A taste for umami . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99
Bernd Lindemann
SEE ARTICLE, PAGE 113

Presynaptic facilitation by hyperpolarization-activated pacemaker channels. . . . 101


Steven A. Siegelbaum
SEE ARTICLE, PAGE 133

Neurodegeneration in the polyglutamine diseases: Act 1, Scene 1 . . . . . . . . . . . 103


Robert Nussbaum and Georg Auburger
SEE ARTICLE, PAGE 157

Non-invasive visualization of cortical columns by fMRI . . . . . . . . . . . . . . . . . . . . 105


Amiram Grinvald, Hamutal Slovin and Ivo Vanzetta
Expertise recruits
SEE ARTICLE, PAGE 164
face-selective brain areas.
Page 191.

brief communications
Rapid, synaptically driven increases in the intrinsic excitability of
cerebellar deep nuclear neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
CD Aizenman and DJ Linden

A taste receptor for


monosodium glutamate.
Pages 99 and 113.

Nature Neuroscience (ISSN 1097-6256) is published monthly by Nature America Inc., headquartered at 345 Park Avenue South, New York, NY 10010-1707. Editorial Office: 345 Park
Avenue South, New York, NY 10010. Telephone 212 726 9200, Fax (212) 696 9635. North American Advertising: Nature Neuroscience, 345 Park Avenue South, New York, NY 10010-
1707. Telephone (212) 726-9200. Fax (212) 696-9006. European Advertising: Nature Neuroscience, Porters South, Crinan Street, London N1 9SQ. Telephone (0171) 833 4000. Fax
(0171) 843 4596. New subscriptions, renewals, changes of address, back issues, and all customer service questions in North America should be addressed to Nature Neuro-
science Subscription Department, PO Box 5054, Brentwood, TN 37024-5054. Telephone (800) 524-0384, Direct Dial (615) 377 3322, Fax (615) 377 0525. Outside North America:
Nature Neuroscience, Macmillan Magazines Ltd., Houndsmill, Brunel Road, Basingstoke, RG21 6XS, U.K.. Tel: +44-(0)1256-329242. Fax: +44-(0)1256 812358. Email:
subscriptions@nature.com. Annual subscription rates: U.S./Canada: U.S. $595, Canada add 7% for GST (institutional/corporate), U.S. $195, Canada add 7% for GST (individual
making personal payment BN: 14091 1595 RT); U.K./Europe:435 (institutional/corporate), 185 (individual making personal payment), 99 (student); Rest of world (excluding
Japan): 480 (institutional/corporate), 195 (individual making personal payment), 110 (student); Japan: Contact Japan Publications Trading Co. Ltd., 2-1 Sarugaku-cho 1 chome,
Chiyoda-ku, Tokyo 101, Japan, phone (03) 292-3755. Back issues: U.S./Canada, $45, Canada add 7% for GST; Rest of world: surface U.S. $43, air mail U.S. $45. Reprints: Nature
Neuroscience Reprints Department, 345 Park Avenue South, New York, NY 10010-1707. Subscription information is available at the Nature Neuroscience homepage at http://neu-
rosci.nature.com. POSTMASTER: Send address changes to Nature Neuroscience Subscription Department, P.O. Box 5054, Brentwood, TN 37024-5054. Application to mail periodicals
postage rate is paid at New York, NY. Executive Officers of Nature America Inc: Nicholas Byam Shaw, Chairman of the Board; Jan Velterop, President; Edward Valis, Secretary-Trea-
surer. Printed by Publishers Press, Shepherdsville, KY, USA. Copyright 2000 Nature America Inc.

nature neuroscience volume 3 no 2 february 2000 i


2000 Nature America Inc. http://neurosci.nature.com

contents

articles
A metabotropic glutamate receptor variant functions as a taste receptor . . . . . . 113
N Chaudhari, AM Landin and SD Roper
SEE NEWS AND VIEWS, PAGE 99

Identification and characterization of the high-affinity choline transporter . . . . . 120


T Okuda, T Haga, Y Kanai, H Endou, T Ishihara and I Katsura

Receptors with opposing functions are in postsynaptic microdomains


under one presynaptic terminal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
G Tsen, B Williams, P Allaire, YD Zhou, O Ikonomov, I Kondova and MH Jacob

Enhancement of synaptic transmission by cyclic AMP modulation of


Gene regulation in
presynaptic Ih channels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133
spinocerebellar ataxia type 1. V Beaumont and RS Zucker
Pages 103 and 157. SEE NEWS AND VIEWS, PAGE 101
2000 Nature America Inc. http://neurosci.nature.com

Synaptic activity modulates presynaptic excitability . . . . . . . . . . . . . . . . . . . . . . 142


TA Nick and AB Ribera

A new form of long-term depression in the perirhinal cortex. . . . . . . . . . . . . . . . 150


K Cho, N Kemp, J Noel, JP Aggleton, MW Brown and ZI Bashir

Polyglutamine expansion down-regulates specific neuronal genes before


pathologic changes in SCA1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 157
X Lin, B Antalffy, D Kang, HT Orr and HY Zoghbi
SEE NEWS AND VIEWS, PAGE 103

High-resolution mapping of iso-orientation columns by fMRI . . . . . . . . . . . . . . . 164


Psychophysics of DS Kim, TQ Duong and SG Kim
Marroquin patterns. SEE NEWS AND VIEWS, PAGE 105
Page 170.
Dynamics of perceptual oscillations in form vision . . . . . . . . . . . . . . . . . . . . . . . . 170
HR Wilson, B Krupa and F Wilkinson

Motion perception during saccadic eye movements . . . . . . . . . . . . . . . . . . . . . . 177


E Castet and GS Masson

Thermosensory activation of insular cortex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184


AD Craig, K Chen, D Bandy and EM Reiman
Inhibition and excitation at
the same axon terminal. Expertise for cars and birds recruits brain areas involved in face recognition. . . . 191
Page 126. I Gauthier, P Skudlarski, JC Gore and AW Anderson

classified advertising
see back pages

nature neuroscience volume 3 no 2 february 2000 ii


2000 Nature America Inc. http://neurosci.nature.com

editorial

How experts communicate


In Darwins time, it was possible to write a book that was both vapid introductory statements like "Much recent research has
a primary scientific report and a popular bestseller. Today, how- been aimed at understanding synaptic plasticity" are as useless
ever, that seems like a remote ideal1. Not only is it difficult to to nonspecialists as they are to anyone else. Concluding para-
2000 Nature America Inc. http://neurosci.nature.com

communicate scientific ideas to the general public, but scien- graphs present a similar temptation to vagueness; saying "this
tists seem to have increasing difficulty communicating with work provides insights" into some problem is less informative
each other. Even within biology, researchers in different areas of than explaining what those insights were. In fact, there is nothing
specialization are often unable to understand each others mysterious about writing for nonspecialists. The key is to exam-
papers. This is a particular problem for a field such as neuro- ine each sentence for hidden assumptions and unfamiliar con-
science, whose advances have often depended on the exchange cepts, and to ensure that they are clearly explained in a way that
of ideas across disciplines. Nature Neuroscience seeks to encour- minimizes the demands on the readers memory.
age clear writing and to make everything we publish accessi- The problems posed by a poorly written article are greater
ble to as many readers as possible. Yet even if this seems for nonspecialists, but even experts comprehend more easily
uncontroversial in principle, it is surprisingly difficult to if they do not have to waste mental resources on parsing diffi-
achieve in practice. cult sentences. Research in linguistics and cognitive psycholo-
Despite the obvious advantages of communicating clearly, sci- gy shows that sentence structure creates expectations about
entists are often resistant to the suggestion that their articles content and emphasis. Writing that violates these expectations
should be comprehensible to readers outside their own field. For is difficult to read 2. Clear writing reduces the demands on
one thing, there is a tendency to equate plain language with over- working memory by presenting information where readers
simplification. As science becomes more complex, the argument expect to find it. Unfortunately, scientific writing often does
goes, an ever-increasing amount of specialist jargon is required the opposite. One common mistake is to separate the sentences
to describe it precisely. Even if this is true, however, technical ter- subject from its verb with a long clause that contains impor-
minology can be explained, and it need not present an insur- tant information (for example, "An increase in mRNA, which
mountable problem to the scientifically literate reader. A more resulted from transcriptional upregulation by factors binding
important deterrent to clear expressionalthough people are to the AP1 site, was observed"). Because the reader is distract-
less willing to admit itis that plain language, no matter how ed by the need for syntactic closure, material between subject
precise, strikes many scientists as somehow unprofessional. It is and verb receives less attention than it should. The opposite
often seen as a badge of academic credibility to express short sim- problem occurs when unimportant material is placed in a loca-
ple ideas in long ponderous phrases; why else would anyone tion that readers naturally emphasize. Each sentence contains
choose to write a sentence such as "To elucidate these issues, we at least one stress position near the end, at the point when
utilized the caprine model" instead of "We studied these ques- readers comprehend how the various parts of the sentence
tions using goats"? relate to each other. Indeed, behavioral studies indicate that
This type of pomposity is easy to avoid once it is recognized, readers slow down as they reach the end of a sentence or
and fortunately many other common problems in scientific writ- clause3. Material at this location is perceived as being impor-
ing are similarly easy to correct. One of the most obvious is exces- tantwhether the author intended it to be or not. Thus, read-
sive use of abbreviations. People within the field are likely to be ers are most comfortable when familiar information at the
familiar with common abbreviations and process them as if they beginning of the sentence creates a context for important new
were words. However, every unfamiliar abbreviation makes an information introduced at the end. These rules do not require
additional demand on the readers memory. Individually, such writers to avoid complicated ideas or long sentences, only to
problems are minor nuisances, but as they accumulate, they can construct them carefully.
severely impair understanding. Another common barrier to com- Because young scientists learn by imitating their elders, a cul-
munication is to describe experimental results in ways that ture of bad writing tends to be self-perpetuating. Perhaps the
emphasize the method of analysis rather than the phenomenon solution is for graduate programs to place more emphasis on for-
being studied. For example, "ANOVA revealed a significant main mal instruction in scientific writing, but this will only happen if
effect of age and a significant interaction effect" is much less infor- scientists appreciate the need for better communication and
mative than "Protein levels decreased significantly with age, and understand the steps that can be taken to achieve it.
this decline was more pronounced in adrenalectomized animals."
1 Gould, S. J. Science 286, 899 (1999).
Even when making the effort to write for a wide readership, 2 Gopen, G. D. & Swan, J. A. Am. Scientist 78, 550558 (1990).
many authors adopt solutions that are ineffective. For example, 3 Just, M. A. & Carpenter, P. A. Psych. Rev. 87, 329354 (1980).

nature neuroscience volume 3 no 2 february 2000 97


2000 Nature America Inc. http://neurosci.nature.com

news and views

A taste for umami


Bernd Lindemann

Chaudhari and colleagues identify the taste receptor for L-glutamate, also known as umami,
found in protein-rich foods. The protein they describe is a new G-protein-coupled receptor that
corresponds to a truncated form of the metabotropic glutamate receptor mGluR4.

Molecular biologist have been trying for ity of taste receptor cells directly, either by downregulating the second messenger
many years to clone taste receptors and contributing to current flow through ion cAMP. This receptor is expressed on
identify their ligands. Now at last, they channels or by modulating channel con- presynaptic terminals of both gluta-
seem to have succeeded. Chaudhari and ductances4. In contrast, sweet, bitter and matergic and GABAergic neurons, where
colleagues have identified a receptor for umami are all thought to depend on the it mediates glutamate-dependent regu-
umami, the taste corresponding to L-glu- activation of G-protein-coupled receptors lation of neurotransmitter release7. In
tamate1. The umami receptor is a G-pro- (GPCRs), but until now, none of these addition, mGluR4 is expressed in taste
tein-coupled receptor, which binds receptors has been definitively identified. tissue, making it an obvious candidate
2000 Nature America Inc. http://neurosci.nature.com

extracellular glutamate and signals for the umami receptor. Howev-


through a G protein to regulate the er, one problem with this idea is
level of intracellular cAMP and the that glutamate activates mGluR4
firing of taste receptor cells. at micromolar concentrations,
Until recently, the textbook wis- far below the taste threshold.
dom was that humans could detect Chaudhari and colleagues
four primary tastes: sweet, bitter, have now resolved this puzzle by
salty and sour. Yet as early as 1908, showing that rat taste tissue also
Kikunae Ikeda at the Tokyo Imperi- expresses an alternative tran-
al University had identified L-gluta- script of mGluR4 (which may
mate as the principal source of a fifth arise by differential transcription
taste. This taste quality, which can- and/or splicing), in which the
not be mimicked by any combina- first 300 amino acids of the
tion of the other four tastes, Ikeda amino (N) terminus are absent.
called umami. Taste researchers have They go on to show that this iso-
long been aware of Ikedas work, but form, when expressed in a cul-
it is only recently that umami has tured cell line, can transduce a
gained widespread recognition in the response to extracellular gluta-
West, perhaps in part because of the mate, over a concentration range
increasing popularity of oriental that is consistent with it being
food 2. Monosodium glutamate the umami taste receptor.
(MSG), of course, is widely used as The loss of the N terminus
a flavoring additive in Asian cuisine, would be predicted to decrease
but the most abundant amino acid, the receptors affinity for gluta-
glutamate, is also an important mate. The sequence of mGluR4 is
Image courtesy of Umami Information Center, Tokyo
nutrient, and it is presumably for this homologous to that of bacterial
reason that some animals have Fig. 1. Umami is an important flavor constituent of many amino-acid binding proteins, for
evolved the ability to taste it. Free protein-rich foods. which crystal structures are avail-
glutamate is found in many protein- able. Based on this homology, the
rich foods, including meat, milk and A report last year described two new N terminus of mGluR4 is predicted to
seafood (Fig. 1); it is particularly concen- GPCRs, TR1 and TR2, that are expressed form a clamshell structure, in which the
trated in aged cheese. Rats and humans can in taste buds. They were suggested to be two halves of the shell are connected by a
both recognize the taste of glutamate, and candidates for sweet and bitter receptors, hinge, forming a cleft in which glutamate
for adult humans the detection threshold but at present there is no functional evi- can bind 810 (Fig. 2). The high-affinity
is about 0.7 mM (ref. 3). dence for this proposal . 5,6 binding site has been mapped by site-
The key to further advance in under- Chaudhari and colleagues1 have pro- directed mutagenesis of the mGluR4
standing taste perception will be the iden- vided such evidence for what seems to sequence, with the hydroxyl groups of Ser
tification of taste receptors. Salty and sour be the umami receptor. The molecule 159 and Thr 182 predicted to form
tastes are produced by small cations such they describe is a new GPCR that corre- hydrogen bonds with glutamate (or its
as Na+, K+ and H+, which affect the activ- sponds to a truncated form of the agonists) and Arg 78 to provide electro-
metabotropic glutamate receptor static attraction. These highly conserved
Bernd Lindemann is in the Department of mGluR4, which they term taste- residues lie within the first half of the
Physiology, Bldg 58, Saar University, D-66421 mGluR4. The mGluR4 receptor was shell structure, and mutating any of them
Homburg, Germany. originally described in the brain, where to alanine causes a major (over 95%)
e-mail: phblin@med-rz.uni-sb.de it responds to extracellular glutamate by reduction in glutamate binding10.

nature neuroscience volume 3 no 2 february 2000 99


2000 Nature America Inc. http://neurosci.nature.com

news and views

A skeptic might still argue that not the transient depolarization. This
taste-mGluR4 is not a taste recep- suggests a model in which glutamate
tor but is instead involved in triggers a decrease in cAMP, resulting in
synaptic transmission or some the closure of cyclic nucleotide-gated
other function of taste tissue, but channels13 and hyperpolarization of taste
several further lines of evidence receptor cells. This would be analogous
are consistent with a role in taste to visual transduction, in which photons
transduction. First, the authors trigger the breakdown of cGMP, result-
confirm that taste-mGluR4 is acti- ing in closure of cGMP-gated cation
vated by the glutamate agonist L- channels and hyperpolarization of pho-
AP4, which to rats is toreceptors. In the case of taste receptors,
indistinguishable from glutamate it is still not known whether hyperpolar-
Fig. 2. The mGluR4 protein is a seven-transmembrane itself11. As with glutamate, the ization modulates tonic release of trans-
receptor, and the N terminus of the brain isoform (center)
concentrations of L-AP4 required mitter, as it does in vertebrate
forms a 'clamshell' structure, homologous to bacterial
amino-acid-binding proteins (left), which contains the high-
to produce a cAMP response are photoreceptors, or whether it has some
affinity binding site for glutamate. The taste isoform (right) consistent with the known taste other effect such as inhibiting the
is truncated at the N terminus. thresholds in rats. Second, the response to other tastants.
same group has previously An additional point of interest is that a
2000 Nature America Inc. http://neurosci.nature.com

shown by in situ hybridization small number of taste cells from the ante-
that mGluR4 expression in taste rior part of the rat tongue have been found
Given the importance of these N-ter- tissue is concentrated in the taste buds to respond to L-AP4 with a depolarization
minal residues, which are absent in the themselves, and that about 40% of buds are rather than a hyperpolarization14. The basis
taste isoform, how can the truncated taste positive for the label. Although the probe for this difference is unknown; perhaps
variant of mGluR4 recognize glutamate used in that study did not distinguish they express channels that are closed rather
at all? Presumably it must contain an between the two isoforms, which may both than opened by cAMP15. The role of these
additional binding site, of lower affinity be present in taste tissue, it is significant cells in taste transduction is also unknown,
than the one at the N terminus. It is that neither isoform was detected outside but it is possible that they, rather than the
unclear whether a similar site exists in the the taste buds, either by in situ hybridiza- hyperpolarizing cells, may be the real
brain isoform, but it is interesting to note tion or by RT-PCR assays11. umami receptors.
that one of the bacterial amino-acid- Finally, the effect of mGluR4 activation Even though it cannot clarify all these
binding proteins was proposed to contain in cultured cells is to decrease cAMP lev- issues, the paper by Chaudhari and col-
a second binding site on the opposite side els, and glutamate is known to reduce leagues is an important advance. The
of the cleft from the first site8. It remains cAMP in isolated taste buds (X. Zhou & N. cloning of a taste receptor with an iden-
to be determined whether glutamate Chaudhari, Chem. Senses 22, 834, 1997). tified ligand should add meat to the field
binds to the taste mGluR4 isoform at the Although the experiments on taste buds of taste research. Bon apptit!
corresponding site or at some other posi- were performed under somewhat artificial
tion, either in the extracellular domains conditionsthe baseline concentration of 1. Chaudhari, N., Landin, A. M. & Roper, S. D.
Nat. Neurosci. 3, 113119 (2000).
or in the bundle of seven helices that span cAMP was raised by treatment with
the membrane. forskolinthey nevertheless seem to reflect 2. Umami Company Report. Umami
Information Center, 1-15-1 Kyobashi, Chuo-
By expressing the two isoforms in a the normal process of taste transduction. ku, Tokyo 104, Japan (1985).
cultured cell line, Chaudhari and col- For instance, the effect of glutamate was 3. Yamaguchi, S. Physiol. Behav. 49, 833841
leagues1 show that activation of the taste enhanced by inositol monophosphate, (1991).
isoform (as measured by the reduction in which, like other ribonucleotides that are 4. Lindemann, B. Physiol. Rev. 76, 719766
intracellular cAMP) requires a much found in meat and other umami foods, (1996).
higher concentration of glutamate than enhances the taste of glutamate24. 5. Hoon, M. A. et al. Cell 96, 541551 (1999).
does the brain isoform. This was not due How does the decrease in cAMP 6. Lindemann, B. Nat. Med. 5, 381382 (1999).
to any difference in the level of expression modulate membrane potential and cause 7. Bradley, S. R. et al. J. Comp. Neurol. 407, 3346
of the two isoforms on the cell surface, so the taste receptor cell to signal the pres- (1999).
it seems likely to reflect a reduced affini- ence of ligand? Electrophysiological 8. Sack, J. S., Saper, M. A. & Quiocho, F. A.
ty for glutamate. This would be consis- recordings of isolated taste cells from the J. Mol. Biol. 206, 171191 (1989).
tent with the truncation of the presumed posterior part of the tongue have shown 9. OHara, P. J. et al. Neuron 11, 4152 (1993).
high-affinity binding site, but it will be two types of responses to L-glutamate12. 10. Hampson, D. R. et al. J. Biol. Chem. 274,
important to confirm the difference in Most cells (60%) respond with a sus- 3348833495 (1999).
affinities by direct binding measurements. tained hyperpolarization, possibly due to 11. Chaudhari, N. et al. J. Neurosci. 16, 38173826
Certainly, it would make sense for the closure of nonselective cation channels. (1996).
umami receptor to have a lower affinity, A few cells (4%) respond with a transient 12. Bigiani, A., Delay, R. J., Chaudhari, N.,
Kinnamon, S. C. & Roper, S. D.
given the high concentrations of gluta- depolarization, probably due to the J. Neurophysiol. 77, 30483059 (1997).
mate that exist in certain foods. The con- opening of such channels.
13. Misaka, T. et al. J. Biol. Chem. 272,
centrations required to activate the It seems likely that the sustained 2262322629 (1997).
receptor are also in the same range as the hyperpolarizing response is what leads to 14. Lin, W. & Kinnamon, S. C. J. Neurophysiol. 82,
known behavioral detection threshold taste signaling, because L-AP4, an ago- 20612069 (1999).
(about 100 micromolar in juvenile and 1 nist that also evokes the taste of umami, 15. Kolesnikov, S. S. & Margolskee, R. F. Nature
mM in adult rodents). also caused the sustained response but 376, 8588 (1995).

100 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

news and views

the K + channel signature sequence


Presynaptic facilitation by (glytyrgly). In addition, the intracellu-
lar C terminus contains a 120-amino-acid
hyperpolarization-activated sequence that is homologous to the cAMP-
and cGMP-binding domains of other pro-

pacemaker channels teins, and is thus the likely site for cAMP
regulation of channel opening8. All four
mammalian genes are expressed in brain,
Steven A. Siegelbaum with differing expression patterns.
In neurons, Ih channels have diverse
Hyperpolarization-activated potassium channels presynaptically functions. They were initially shown to be
facilitate transmitter release in a calcium-independent fashion. inward rectifiers; they are active near the
resting potential and pass inward current
The past few years have seen rapid unusual property of these channels is their more readily than outward current, there-
advances in our understanding and appre- regulation by cyclic nucleotides1, which by helping to control resting potential and
ciation of hyperpolarization-activated non- speed the rate of channel activation by input resistance2. In photoreceptors, Ih
selective cation channels, also known as Ih binding to an intracellular site on the chan- channels help to damp the hyperpolarizing
channels. Although the first well-charac- nel (Fig. 1b). In the heart, this speeds the effect of light; they are activated by hyper-
2000 Nature America Inc. http://neurosci.nature.com

terized role of these channels was the con- slow phase of spontaneous depolarization polarization, causing the voltage response
trol of pacemaking activity in sinoatrial (and hence the heart rate) in response to to light to fade during the first 100200 ms,
node cells of the heart1, they are also wide- -adrenergic receptor stimulation. thus producing sensory adaptation. In many
ly expressed in peripheral and central neu- The genes encoding Ih channels were CNS neurons, activation of Ih channels fol-
rons 2. Neuronal I h channels control recently cloned from both mammals46 and lowing inhibitory postsynaptic potentials
rhythmic electrical activity in sponta- sea urchins7 (see ref. 8 for review). These contributes to a rebound afterdepolariza-
neously active cells and regulate membrane genes, called HCN14 in mammals, are tion (ADP), which can trigger an action
excitability in quiescent cells. On page 133 members of the voltage-gated K+ channel potential. Ih can also contribute to sponta-
of this issue, Beaumont and Zucker3 report family. The encoded proteins contain six neous activity in neurons, similar to its role
a new role for Ih channelsthey contribute transmembrane segments, including a pos- in the heart. For instance, in thalamocortical
to presynaptic facilitation of transmitter itively charged S4 voltage sensor (Fig. 1a) relay neurons, activation of Ih underlies a
release. Surprisingly, this facilitation does and a pore-forming P region that includes burst firing pattern associated with slow-
not seem to involve an increase in intracel-
lular calcium; instead, the authors raise the a P
b
provocative possibility that it may involve a out -50 mV
direct coupling between these channels and s1 s2 s3 s4 s5 s6
the vesicular release machinery. in -90 mV
Ih channels have several distinctive fea-
tures. Unlike most voltage-gated channels, CNBD
they open in response to negative-going Control
voltage steps to potentials within the range
+cAMP
of the normal resting potential (Fig. 1b). N C
They conduct both potassium (K+) and d 50 Hz
control
sodium (Na+) ions, with a three-fold greater
prox
permeability to K +, yielding a reversal
potential of 30 to 40 mV under physio- c +cAMP
dist

logical conditions. As a result, the opening +40 mV


Control
of Ih channels near the resting potential
(~ 60 mV) generates an inward, depolar- 10 mV
izing current that is largely carried by Na+. +ZD7288
50 ms
In heart pacemaker cells, these channels -60 mV
dist
contribute to the slow phase of depolariza- prox

tion that follows the repolarization phase Ih ICa,T


of the action potential. As the membrane Amy Center

potential reaches threshold, voltage-gated Fig. 1. Ih channels and their role in excitability. (a) Schematic of transmembrane topology of the
Na+ channels and T-type Ca2+ channels are HCN hyperpolarization-activated channels. (b) Hyperpolarizing voltage step from 50 mV to 90
activated, generating the rapid rising phase mV activates an inward Ih current with slow kinetics. On returning to the holding potential, there is
of the next action potential, during which an inward tail current as the Ih channels deactivate. Activation of Ih under basal conditions (blue)
the Ih channels shut (Fig. 1c). Another and after elevation of cAMP (red). (c) Contribution of Ih current to spontaneous action potential
generation under basal conditions (blue) and after elevation of cAMP (red). (d) Role of Ih in den-
dritic integration. EPSPs recorded in cell body of hippocampal CA1 pyramidal neuron in response
Steven Siegelbaum is at the Center for Neurobiology, to stimulation of distal or proximal Schaffer collateral inputs. Top traces, control. Bottom traces,
Department of Pharmacology, Howard Hughes after blockade of Ih with ZD7288 (from ref. 9). Ih channels, localized at a high density in the distal
Medical Institute, Columbia University, 722 W. 168 dendrite, normally act to speed the rate of decay of distal EPSPS, perhaps by decreasing the local
St., New York, New York 10032, USA. membrane time constant. On blockade of Ih channels, there is a preferential slowing of the decay
e-mail: sas8@columbia.edu phase of the distal EPSP, which increases temporal summation.

nature neuroscience volume 3 no 2 february 2000 101


2000 Nature America Inc. http://neurosci.nature.com

news and views

wave sleep. Regulation of Ih in these cells by ing, given that they have been detected elec- (Fig. 2b). However, a previous study from
cAMP is important in the sleepwake cycle2. trophysiologically in the large presynaptic the Zucker lab using Ca2+ indicator dyes
Ih channels are also expressed in dendrites, terminals of the chick giant calyx10 and by failed to detect any increases in Ca2+ levels
where they influence the cable properties of antibody staining on the basket cell termi- in response to 5-HT at crayfish presynap-
the dendrite and shape the time course of nals surrounding cerebellar Purkinje neuron tic terminals13. Instead, measurements of
the EPSP as it is propagated to the soma9 soma4 (Fig. 2a). What is new about the pre- synaptic vesicle cycling with the membrane
(Fig. 1d). sent study is their proposed function and the dye FM1-43 from the same group14 showed
In their new study, Beaumont and unusual mechanism by which these chan- that 5-HT increases the pool of readily
Zucker3 extend the role of Ih in neurons by nels regulate synaptic transmission. releasable vesicles.
showing that these channels can alter neu- Previous studies on the crayfish neuro- To explain their findings, Beaumont and
rotransmitter release from presynaptic ter- muscular junction have shown that the Zucker suggest a provocative hypothesis in
minals. Recording from the motor terminals neurotransmitter serotonin (5-HT), acting which opening of Ih channels enhances the
at crayfish neuromuscular synapses, the through both cAMP and phosphatidyl mobilization of synaptic vesicles to a readi-
authors demonstrate a pronounced ADP fol- inositol second-messenger pathways, caus- ly releasable pool (Fig. 2). The authors sug-
lowing a hyperpolarizing voltage step, a hall- es a facilitation of glutamate release from gest that this effect could be due to a direct
mark of Ih activation. To confirm that the excitatory presynaptic terminals11. Beau- interaction of Ih channels with the release
ADP is indeed due to Ih, the authors show mont and Zucker now show that the effect machinery, perhaps mediated by the
that it is blocked by inorganic and organic of 5-HT depends, at least in part, on the cytoskeleton. This idea is not without prece-
2000 Nature America Inc. http://neurosci.nature.com

blockers of Ih. The presence of Ih channels at direct activation of presynaptic Ih by cAMP. dent, in that voltage-gated calcium channels
these terminals is not particularly surpris- Application of 5-HT, or direct elevation of interact directly with the presynaptic
cAMP by forskolin, depolar- SNARE protein syntaxin, regulating both
izes the presynaptic terminal channel function and vesicle fusion15. Alter-
by 510 mV and enhances the natively, increased influx of Na+ through
magnitude of the postsynap- activated Ih channels could influence some
tic response by potentiating local signaling cascade. Whatever the mech-
transmitter release. Although anism, it seems to involve the stable activa-
cAMP often works by activat- tion of some downstream signal; the authors
ing protein kinase A (PKA), show that direct activation of Ih (by a one-
its effect at these terminals minute hyperpolarization of the presynap-
could not be attributed to tic axon) is sufficient to induce an
PKA, suggesting that cAMP enhancement of transmitter release that per-
may instead activate some sists for as long as 20 minutes. Although fur-
other target protein by a direct ther work is needed to pin down this
mechanism. Because Ih is acti- mechanism, the new findings clearly point
vated directly by cAMP, it was to an unforeseen role for Ih in the control of
a plausible candidate, and the neuronal function.
authors confirmed its role by
showing that pharmacologi- 1. DiFrancesco, D. Annu. Rev. Physiol. 55, 455472
(1993).
cal agents that inhibit Ih par-
2. Pape, H. C. Annu. Rev. Physiol. 58, 299327 (1996).
tially block the ability of 5-HT
3. Beaumont, V. & Zucker, R. S. Nat. Neurosci. 3,
or forskolin to depolarize the 133141 (2000).
presynaptic terminal and
4. Santoro, B., Grant, S. G., Bartsch, D. & Kandel,
enhance transmitter release. E. R. Proc. Natl. Acad. Sci. USA 94, 1481514820
How might activation of Ih (1997).
facilitate release? One possi- 5. Santoro, B. et al. Cell 93, 717729 (1998).
bility is that it could enhance 6. Ludwig, A., Zong, X., Jeglitsch, M., Hofmann, F.
Ca2+ influx into the presynap- & Biel, M. Nature 393, 587591 (1998).
tic terminal, for example by 7. Gauss, R., Seifert, R. & Kaupp, U. B. Nature 393,
causing depolarization and 583587 (1998).
Bob Crimi
thus opening voltage-gated 8. Santoro, B. & Tibbs, G. R. Ann. NY Acad. Sci.
2+ 868, 741764 (1999).
Fig. 2. Models for presynaptic facilitation (a, b) Presynaptic Ca channels or by increasing
the rate of presynaptic action 9. Magee, J. C. Nat. Neurosci. 2, 508514 (1999).
terminal without (a) and with (b) 5-HT, showing different
modes of regulation of transmitter release during cAMP- potential firing. There is a 10. Fletcher, G. H. & Chiappinelli, V. A. Brain Res.
dependent presynaptic facilitation. Closure of K+ channels in precedent for this in the mol- 575, 103112 (1992).
Aplysia neurons, via phosphorylation by protein kinase A, lusk Aplysia, where 5-HT (act- 11. Dixon, D. & Atwood, H. L. J. Neurophysiol. 62,
broadens the action potential and increases Ca2+ influx ing through PKA- and 12511259 (1989).
through voltage-gated calcium channels, which enhances PKC-dependent pathways) 12. Byrne, J. H. & Kandel, E. R. J. Neurosci. 16,
release. There is also a more direct effect of protein kinase A enhances transmitter release 425435 (1996).
phosphorylation on the release machinery. At the crayfish 13. Delaney, K., Tank, D. W. & Zucker, R. S.
at a sensorimotor synapse by J. Neurosci. 11, 26312643 (1991).
neuromuscular junction presynaptic terminals, 5-HT acts in a +
Ca -independent manner to enhance the pool of readily closing presynaptic K chan-
2+
14. Wang, C. & Zucker, R. S. Neuron 21, 155167
releasable synaptic vesicles. Modulation of Ih contributes to nels, thereby prolonging the (1998).
this effect by an unknown mechanism that does not involve presynaptic action potential 15. Catterall, W. A. Ann. NY Acad. Sci. 868, 144159
enhanced Ca2+ influx. and enhancing Ca2+ influx12 (1999).

102 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

news and views

approach, they were able to identify tran-


Neurodegeneration in the scripts that are either upregulated or
downregulated in the mutant mice. The
polyglutamine diseases: authors identified seven such genes, and
for each one they determined when in

Act 1, Scene 1 development expression first begins to


differ between mutant and wild-type
mice. Surprisingly, one transcript was
Robert Nussbaum and Georg Auburger found to be downregulated on postnatal
day 11, only one day after the Purkinje-
A mouse model of the neurodegenerative disease cell specific promoter begins to drive
spinocerebellar ataxia type 1 reveals that changes in gene expression of the ataxin-1 transgene, and
expression begin many weeks before the onset of symptoms. at least 56 weeks before the first mani-
festations of the disease. This transcript
encodes prenylcysteine carboxylmethyl-
The best therapy for neurodegenerative defined genetic causes that lead repro- transferase (PCCMT), an enzyme
diseases is likely to be prevention, given ducibly to a common endpointdeath enriched in the endoplasmic reticulum
the limited regenerative capacity of cen- of neurons by apoptosisthe polygluta- (ER) of the cerebellum, which partici-
2000 Nature America Inc. http://neurosci.nature.com

tral nervous tissue. To design rational mine disorders provide a powerful pates in post-translational lipid modifi-
approaches for the prevention of these model in which to study the pathogen- cation of many proteins, including the
diseases, therefore, we need to identify esis of neurodegeneration. G protein RAS. Later on, but still 23
the initial pathological events and SCA1 is characterized by the onset weeks before the onset of clinical signs
understand how they ultimately lead to (usually in adulthood) of cerebellar and or Purkinje cell pathology, transcripts
neuronal cell death. In this issue of bulbar dysfunction. This is due to a encoding proteins with effects on cellu-
Nature Neuroscience, Lin et al.1 provide severe loss of cerebellar Purkinje cells as lar calcium and glutamate metabolism
an enticing glimpse of the first cellular well as atrophy and degeneration in var- were also downregulated. These includ-
changes in the hereditary neurodegen- ious other regions of the brain and spinal ed the type I ER inositol triphosphate
erative disease spinocerebellar ataxia cord. The disease has been studied over receptor IP3R1, inositol polyphosphate
type 1 (SCA1). They have used a mouse a number of years, notably in the labo- 5-phosphatase (INPP5A), an ER calcium
model to identify genes whose expres- ratories of Huda Zoghbi and Harry Orr, pump (SERCA2), the calcium ion chan-
sion is altered in the earliest stages of the who in 1993 identified ataxin-1 as the nel TRP3 and the glutamate transporter
disease; moreover, their preliminary gene carrying the pathological polyglut- EAAT4, which is located in dendritic
results suggest that many of these genes amine expansion 3. Although the wild- spines of Purkinje cells. Only later, when
show similar alterations in human SCA1 type ataxin-1 protein has no known pathological and clinical signs first start-
patients. function, the mutant form gives rise to ed to appear, did another gene begin to
SCA1 is one of eight hereditary neu- aggregates that accumulate in the nuclei be upregulated; that gene was the early-
ropathies (the best-known being Hunt- of affected neurons. Perhaps surprising- stage acute phase protein and AD aggre-
ingtons disease) that are caused by the ly, ataxin-1 expression is not confined to gation marker alpha1-antichymotrypsin.
pathological expansion of a trinucleotide the cells that die in the disease, but is Importantly, these changes (apart from
repeat that encodes polyglutamine2. The ubiquitous in both the nervous system the upregulation of alpha1-antichy-
unraveling of the mechanism underly- and other tissues. Thus, the expression motrypsin) were only seen in mice
ing these disorders has been one of the pattern alone cannot explain the charac- expressing the expanded ataxin-1 in the
great success stories of the last decade. teristic neuronal degeneration observed nucleus; no change was seen in control
Although each of the diseases involves a in SCA1. Zoghbi, Orr and colleagues mice expressing either wild-type ataxin-1
different gene and different clinical and went on to develop transgenic mouse or an expanded ataxin-1 that lacked a
neuropathological features, the under- models for SCA1, using a Purkinje-cell nuclear localization domain and was
lying mechanism is the same in each specific promoter to drive the expression confined to the cytoplasm.
case; a piece of DNA consisting of CAG of various forms of mutant ataxin-1, and What about human SCA1 patients?
repeats becomes amplified, leading to a created mice with cerebellar degenera- Although it is difficult to obtain materi-
protein product that contains a patho- tion similar to that found in human al for RNA studies, the authors were able
logically expanded string of glutamine patients. By expressing mutant ataxin-1 to confirm by immunohistochemistry
residues. These disorders are relatively that lacked either a nuclear localization that three of the gene products down-
rare compared to (say) Alzheimers or signal or an aggregation domain, the regulated in mutant mice (PCCMT,
Parkinsons disease, but their significance authors were able to show that nuclear SERCA2 and IP3R1) also showed
goes beyond their impact on public localization of abnormal ataxin-1, and decreased expression in the brain of an
health. Because they arise from well- not aggregation per se, was sufficient to early-onset SCA1 patient, relative to age-
cause the phenotype4. matched controls. Alpha1-antichy-
The authors are at the National Human In the new paper 1 by Lin et al., the motrypsin, which is upregulated in the
Genome Research Institute, National Institutes same groups continue their analysis of mutant mice, was also upregulated in the
of Health, 49 Convent Drive, Bethesda, transgenic mouse models for SCA1 to human patient. Consistent with the con-
Maryland 20892, USA. identify the earliest changes in gene clusions of Lin et al.1, unpublished work
email: rlnuss@nhgri.nih.gov or expression in the cerebellum. By using a from one of our laboratories (G.A.) has
auburger@nhgri.nih.gov PCR-based subtractive hybridization indicated that a number of transcripts

nature neuroscience volume 3 no 2 february 2000 103


2000 Nature America Inc. http://neurosci.nature.com

news and views

ronal death later in extends beyond the polyglutamine dis-


the disease process, orders. Various abnormal cellular
and if so, by what processes have been proposed as the first
mechanism? Here we steps in the eventual neuronal loss
can only speculate. observed in all of the major neurode-
PCCMT is involved generative diseases, including both
in the post-transla- Alzheimers and Parkinsons disease.
tional modification Among the leading candidates are pro-
of many proteins, tein aggregation, damage from oxygen
any one of which free radicals (oxidative stress)8 and exci-
might be involved in totoxicity, any of which, alone or in
neuronal degenera- combination, could result in the induc-
tion. The other tion of apoptosis. Which of these are late
downregulated genes phenomena, and which are important at
are involved in the earliest stages of disease? In princi-
regulating cytoplas- ple, these questions can be addressed in
mic calcium and glu- carefully staged human brain tissue, but
tamate neurotrans in practice such material is not readily
2000 Nature America Inc. http://neurosci.nature.com

mission; IP3R1 stim- available, meaning that mutant mice of


ulates intracellular the type used by Lin et al. are likely to be
Ca2+ release, INPP5A central in future advances.
may act to terminate Although the implications of this
the biological activity study must still be considered tentative,
of IP3, SERCA2 and it is striking that so many of the early
TRP3 are both changes seen by Lin et al. are consistent
involved in calcium with an effect on excitotoxicity, whereas
flux across mem- none seems to implicate oxidative stress
branes, and EAAT4 is as an early event in the SCA1 mouse
involved in clearing model. The notion that dysregulation of
excess glutamate glutamate and calcium signaling is a very
from the extracellular early event in the progression of neu-
space. One plausible rodegeneration will, if substantiated by
hypothesis is that future work, stimulate the design of
downregulation of therapeutic approaches that may even-
Fig. 1. Pre- and postsynaptic neurons, with intervening synaptic cleft. these genes might tually make a difference to the clinical
Shown are some of the molecules implicated by transcript profiling in promote excitotoxici- management of many neurodegenera-
the earliest stages following expression of pathologically expanded ty, a well-character- tive conditions. Indeed, a number of
ataxin-1 (courtesy of D. Leja). ized phenomenon in pharmacological compounds that influ-
which neurons die by ence calcium and glutamate homeosta-
apoptosis due to over- sis are already available, and one such
excitation of gluta- compound, riluzole, has proven to be
involved in regulating calcium, inositol mate receptors and a consequent increase the most effective therapy so far in
and glutamate neurotransmission show in cytoplasmic calcium5. Consistent with patients with amyotrophic lateral scle-
altered expression in the striatum of this idea, many of the neurons affected rosis, a neurodegenerative disease of the
presymptomatic and early-stage Hunt- in the different polyglutamine disorders spinal cord with an entirely different eti-
ingtons disease patients. show strong staining with various calci- ology than the polyglutamine disorders9.
The findings of Lin et al.1 raise two um-regulating proteins and receive glu- 1. Lin X., Antalffy, B., Kang, D., Orr, H. T. &
fundamental questions that must be tamatergic synaptic input 6. Moreover, Zoghbi, H. Y. Nat. Neurosci. 3, 157163
answered before the pathogenesis of excessive activation of NMDA-type glu- (2000).
SCA1 can be understood. First, we want tamate receptors can produce a clinical 2. Lunkes, A., Trottier, Y. & Mandel, J. L. Essays
to know how the mutant ataxin leads to and neuropathological pattern reminis- Biochem. 33, 149163 (1999).
altered gene expression. The possibili- cent of Huntingtons disease in animals7, 3. Orr, H. T. et al. Nat. Genet. 4, 221226
(1993).
ties include a direct effect on the tran- and overstimulation of AMPA-type glu-
scription of specific genes, a more global tamate receptors can trigger the selective 4. Klement, I. A. et al. Cell 95, 4153 (1998).
effect on chromatin structure, nuclear degeneration of Purkinje neurons of the 5. Schwarcz, R., Foster, A. C., French, E. D.,
Whetsell, W. O. Jr. & Kohler, C. Life Sci. 35,
architecture or transcription factor sta- cerebellum. Clearly these issues cannot 1932 (1984).
bility, or a change in the stability of par- be resolved simply by looking at tran- 6. Greenamyre, J. T. Arch. Neurol. 43,
ticular RNA trancripts. With the scriptional profiles, but the availability 10581063 (1986).
availability of a mouse model for the of mouse models should allow more 7. diFiglia, M. Trends Neurosci. 13, 286289
disease, this is likely to become an area direct genetic and physiological tests of (1990).
of intensive inquiry. the excitotoxicity hypothesis. 8. Beal, M. F. Ann. Neurol. 38, 357366 (1995).
Second, are these early changes in As we noted at the outset of this arti- 9. Doble, A. Pharmacol. Ther. 81, 163221
gene expression the actual cause of neu- cle, the significance of these findings (1999).

104 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

news and views

our own optical imaging data showing


Non-invasive visualization of ocular dominance columns in behaving
monkeys (Figs. 1 and 2) and by referring
cortical columns by fMRI to previous work using biophysics and
optical imaging.
Amiram Grinvald, Hamutal Slovin and Ivo Vanzetta We determined the activity-dependent
spatiotemporal characteristics of hemody-
Functional magnetic resonance imaging can now resolve namic events, including oxygen delivery,
blood flow and blood volume, at the onset
individual cortical columns, which should provide insights
of cortical activity triggered by sensory
into sensory perception and higher cognitive functions. stimulation. The first event is a prolonged
increase in oxygen consumption, caused
Mountcastle1 and Hubel and Wiesel2 orig- fMRI, achieving sufficient spatial resolu- by an increase in oxidative metabolism of
inally showed that many complex neural tion to directly visualize cortical columns activated neurons, leading to an increase
networks display meticulous order. Neu- responsible for shape perception in visual in the deoxyhemoglobin concentration
rons with common functional properties cortex of anesthetized cats. Instead of the starting less than 100 ms after activity
are often clustered together in columns, traditional positive BOLD fMRI signal, onset10,11. Upon illumination at 605 nm,
2001000 m wide, which span the depth the authors used an earlier, negative phase this early event causes cortical darkening
2000 Nature America Inc. http://neurosci.nature.com

of the cortex. To discover principles under- of the signal, termed the initial dip (Figs. 1b, middle panel, and 2b) known to
lying implementation of the neural code, (deoxygenation phase; Fig. 1b in ref. 8), be colocalized with the site of electrical
we must understand functional organiza- to obtain single-condition maps of corti- activity in columns (see Fig. 4 in ref. 7).
tion across the sheet of cortical columns cal columns. They further showed that the The second event originates from a delayed
during sensory processing and cognitive traditional BOLD signal (positive hyper- increase in blood volume, starting about
tasks. Brain organization has been visual- oxygenation phase) gives less precise res- 300500 ms later10, caused by capillary
ized3 with positron-emission tomography olution than this early phase. dilation, which also causes cortical dark-
(PET), optical imaging of intrinsic signals Because it remains controversial exact- ening, particularly apparent in larger blood
and, most recently, functional magnetic ly how electrical activity is coupled to such vessels (Figs. 1c and d, middle panel, and 2e).
resonance imaging (fMRI). Optical imag- metabolic responses, skeptics question Blood volume changes occur almost simul-
ing allows detailed visualization of corti- whether imaging maps based on micro- taneously in the three vascular compart-
cal columns (spatial resolution of 20100 circulation responses indeed represent the ments and are not well regulated within
m, verified by extensive single-neuron brains electrical activity or just hemody- individual cortical columns (Fig. 4 in ref. 7).
and histological mapping), but it is inva- namics. Thus methods to distinguish The third event is an activity-related
sive and thus unsuitable for use in between brain and vein9 are critical. After increase in blood flow, as blood rushes into
humans. PET and fMRI offer spectacular Kim and colleagues presented their capillaries. Starting about 0.51.5 seconds
three-dimensional localization of activity results 8 at the 1999 Society for Neuro- after the onset of electrical activity, this
in humans, but have lacked the spatial res- science meeting, some felt that neurosci- decreases deoxyhemoglobin concentration
olution to reveal cortical columns. entists fantasy might soon become reality. and increases oxyhemoglobin concentra-
These techniques depend on the pio- For others, questions remained. Exactly tion. The delayed deoxyhemoglobin
neering finding of Roy and Sherrington4 how is cortical electrical activity related to decrease is much larger than the initial
that changes in electrical activity are cou- various hemodynamic responses? Why increase, causing delayed cortical whiten-
pled to microcirculation responses, whose was the initial dip not observed in most ing, particularly apparent in larger blood
regulation is still being explored. Neural previous studies? What are the implica- vessels (Fig. 1b and c, right). Unlike the
activity locally increases metabolic tions for standard low-resolution fMRI at early deoxygenation phase, this delayed
demands for glucose and oxygen, and the low field strength? Can the impressive sin- increase is not localized with columnar
brain microcirculation respondsmuch gle-condition orientation columns electrical activity. Thus, changes in deoxy-
less locallyby increasing blood volume obtained by fMRI in the anesthetized cat hemoglobin concentration follow a bipha-
and flow. In PET, this enhanced blood also be visualized in awake animals? Are sic time course: a deoxyhemoglobin
flow is visualized by injecting radioactive these results relevant to primates? increase because of increased oxidative
material into the circulation. Blood oxy- The relationship of electrical activity metabolism of active neurons the ini-
genation level-dependent5 (BOLD) fMRI to hemodynamic responses has been tial dip and then a decrease due to large
instead uses concentration changes in addressed by optical imaging 6,7,10,11 . overcompensation by enhanced blood flow
paramagnetic deoxygenated hemoglobin Because the spatial resolution of the optics (Figs. 1b and 2b). Obviously, increased
as an intrinsic contrast agent. In optical is less than 1 m, this method permits oxidative metabolism must localize with
imaging, this contrast agent yields high- unambiguous identification of blood-ves- electrical activity.
resolution maps of cortical columns6,7, sel-derived artifacts, distinguishing them With high-field fMRI, Logothetis and
and BOLD imaging was hoped to have from cortical activation per se. In addition, colleagues12 observed the initial dip in cor-
similar potential. Kim and colleagues 8 photons are cheap, so one can collect tical regions without large vessels but not
now report a striking technical advance in plenty of them; thus the signal-to-noise in large vessels, whereas the late BOLD
ratio is higher than for fMRI. This allows component was detectable in both com-
The authors are in the Department of direct visualization of hemodynamic partments (see Fig. 5 in ref. 12). These
Neurobiology, The Weizmann Institute of changes without elaborate image process- findings support the claims of Kim and
Science, Rehovot 76100, Israel. ing or statistics. Below we address these colleagues8 that the initial fMRI dip shows
e-mail: amiram.grinvald@weizmann.ac.il five remaining questions by presenting better spatial specificity. Thus, cortical

nature neuroscience volume 3 no 2 february 2000 105


2000 Nature America Inc. http://neurosci.nature.com

news and views

spatial resolution. The point spread of the


fMRI signal is large relative to cortical
column diameter14. However, differential
imaging can resolve two distinct cortical
activation sites at distances much smaller
than half the width of the signal spread,
if the signal-to-noise ratio is adequate.
Indeed, ocular dominance columns in
humans have already been imaged with
fMRI by this method 15 (see also T.W.
James et al. Soc. Neurosci. Abstr. 25,
212.5,1999; Tanaka et al., Soc. Neurosci.
Abstr. 25, 572.3, 1999). As discussed
above, both optical imaging and BOLD
fMRI signals have multiple components.
The local component probably origi-
nates from oxygen extraction from the
capillaries; thus it is most closely coupled
2000 Nature America Inc. http://neurosci.nature.com

to electrical activity and most stimulus-


specific. This component is particularly
enhanced during the initial dip, before
delayed hemodynamic events begin to
obscure it. The global signal7,10 refers to
the sum of all the components, which
Fig. 1. Comparison between differential and single-condition maps of ocular dominance columns, originate from this deoxygenation and
visualized by optical imaging. A cranial window was implanted over the primary visual cortex of a other sources, such as nonlocalized
macaque. The exposed cortical surface of the behaving monkey was illuminated at 605 nm to blood-volume and blood-flow changes
emphasize small changes in deoxyhemoglobin concentration. Each row represents a time series, (black and white vessels, respectively, in
taken at roughly two-second intervals beginning one second before stimulus onset. (a) Differential Fig. 1ad, late frames). Most of the inte-
images, obtained by subtracting images obtained with left- versus right-eye stimulation. To show grated global signal is associated with
time development of the differential map, all images are clipped at the same range. (Clipping is these last two components, which may
determining the range and mean of the gray scale's contrast.) (b) Corresponding single-condition not be stimulus-specific as they reflect
maps obtained by normalizing the cortical image to illumination intensity without any additional
hemodynamic activation of venules and
processing, when the contralateral eye viewed the stimulus. To emphasize the slow global changes,
a fixed-range clipping with fixed mean values was used. (c) As in (b), but to emphasize the small large draining veins far from the electrical
changes, a fixed range was used without a fixed mean. (d) As for (b), but to maximize the dynamic activation site7,10. For this reason, when
range of each map, autoclipping was used. Note that the signal-to-noise ratio in a differential image two stimuli are orthogonalmeaning
is much higher than that obtained for a single-condition map. Scale bar, 1 mm. that they activate neurons in comple-
mentary cortical patchescomparing
their activation patterns enhances the sig-
electrical activity is most tightly coupled evidence suggest that the amplitude of the nals local component, while eliminating
to hemodynamic events during the initial initial dip depends on the second power many, though not all, global components.
dip. Because the initial dip is localized of the field strength (K. Ugurbil, personal Therefore, when the assumptions dis-
with neuronal activity, whereas the sub- communication). The existence of the ini- cussed below are fully justified, differen-
sequent larger decrease in deoxyhemo- tial dip has been confirmed by several tial imaging may reveal the correct pattern
globin is not, there is a time window groups using 4 T fMRI in humans13. For of electrical activation. Such maps may
during the first three or four seconds in those debating whether to purchase ultra- even yield a better signal-to-noise ratio
which functional maps indeed correspond expensive high-field magnets, it will be than maps derived from the shorter peri-
to electrical activity. In contrast, delayed critical to establish the relative amplitude od and smaller amplitude of the initial
microvascular events such as volume and and signal-to-noise ratio of the initial dip dip. However, differential imaging has two
flow changes, which are not localized with as a function of field strength. major limitations. First, it works perfect-
electrical activity, may yield hemody- Kim and colleagues reported single- ly only if stimuli are orthogonal in the
namic maps that do not correspond to condition maps (Fig. 2ad in ref. 8). To above sense, such as orthogonal oriented
electrical activation (compare Fig. 2g and understand the implications of their stimuli activating orientation columns in
h with Figs. 3a and b and 4c in ref. 8). advance for standard low-field fMRI cats and monkeys. Even for ocular domi-
Why have low-field (0.51.5 T) BOLD research (the third question), we must nance maps, the stimuli are not strictly
fMRI measurements failed to detect the clarify the advantages and limitations of orthogonal because contralateral activa-
initial dip observed by Kim and colleagues single-condition mapping compared tion is stronger than ipsilateral activation,
with high-field (4.7 T and 9.4 T) magnets with differential mapping (for details, so that blood vessel-derived artifacts may
and by optical imaging? One possibility is see ref. 3, pages 918921). The spatial res- be the largest signals in differential maps
that low-field measurements are not suf- olution of a functional map depends on computed from late hemodynamic
ficiently sensitive to deoxyhemoglobin the spread of a particular imaged signal responses (Fig. 2c and compare early vas-
concentration changes in cortical capil- beyond the site of electrical activity, sig- cular artifacts in Fig. 2f with late ones in
laries. Both theoretical and experimental nal-to-noise ratio and the instruments Fig. 2i). More generally, functional map

106 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

news and views

Finally, skeptics tional technical improvements are still


wondered whether sin- required before high-resolution single-
gle-condition orienta- condition mapping becomes a gold stan-
tion columns obtained dard procedure easily achieved by fMRI.
in anesthetized cats 8 Such improvements might include exten-
can be visualized in sive averaging, use of different MRI
awake animals, and sequences, improved ways to reject vas-
whether these results cular artifacts or reliance on MRI signals
are relevant to pri- other than BOLD.
mates. Our new results The advance described by Kim and
from awake monkey colleagues8 represents a major step for-
cortex (Figs. 1 and 2), ward in functional brain imaging. It
which agree with those should lay the foundation for exploration
of Kim and col- of the human brain at the fundamental
leagues8, address both level of its columnar architecture. This
these concerns. Hence approach may revolutionize fMRI studies
it seems reasonable to by allowing them to address not only
hope that human where but also how processing and com-
2000 Nature America Inc. http://neurosci.nature.com

brain imaging with putations are carried out by individual


fMRI will also allow cortical columns. The combination of
Fig. 2. Comparison between single-condition and differential maps high-resolution single- fMRI with techniques that reveal cortical
obtained during the initial dip (deoxygenation phase) and later rise condition mapping. dynamics in the millisecond time domain,
(hyperoxygenation phase). (a) Cortical image taken in green light to Kim and colleagues such as electroencephalography and mag-
emphasize the vascular pattern. Scale bar, 1 mm. (b) Time course of accomplished the dif- netoencephalography, is also warranted.
the amplitude of the global signal obtained from the single-condition ficult feat of single- In conjunction with such methods, fMRI
maps. (c) Time course of vascular artifacts and of the mapping signal
condition mapping by will become a powerful non-invasive tool
amplitude calculated from the differential map. (df) Maps from the
early deoxygenation phase. (d) Single-condition map for the con- integrating BOLD sig- both in biomedicine and in basic research
tralateral eye. (e) As in (d), for the ipsilateral eye. (f) The differential nals mainly during the of neurophysiology underlying higher
map. (gi) As for (df), from the late period. Note that during this small initial dip. As brain functions, thus bridging the gap
late time, the most prominent signals in the rather poor single-condi- with any new between psychology and neurobiology.
tion maps (g, h) are artifacts from blood vessels (diameter 50150 approach, additional
m). Although these brain or vein artifacts are minimized by differ- research will undoubt- 1. Mountcastle, V. B. J. Neurophysiol. 20, 408434
(1957).
ential mapping (f), at least with optical imaging, they are larger than edly strengthen this
the columnar signal. 2. Hubel, D. H. & Wiesel, T. N. J. Physiol. (Lond.)
works foundations. 160, 106154 (1962).
First, the authors did 3. Windhorst, U. & Johansson, H. (eds) Modern
not present unequivo- Techniques in Neuroscience Research (Springer,
amplitudes can be three- to tenfold small- cal single-neuron confirmation, or con- New York, 1999).
er than ocular dominance column maps firmation by two-deoxyglucose 4. Roy, C. & Sherrington, C. J. Physiol (Lond.). 11,
shown here, making vessel-derived arti- autoradiography or optical imaging, 85-108 (1890).
facts far more problematic. Furthermore, which would be even better. However, the 5. Ogawa, S., Lee, T. M., Kay, A. R. & Tank, D. W.
Proc. Natl. Acad. Sci. USA 87, 98689872
orthogonal stimuli may be impossible to complementary nature of the single-con- (1990).
design, particularly for columns repre- dition maps for the two sets of orthogo-
6. Grinvald, A., Lieke, E., Frostig, R. D, Gilbert, C.
senting higher cognitive functions. Sec- nal orientations strongly suggests that D. & Wiesel, T. N. Nature 324, 361364
ond, by far the largest signals are from their maps were indeed orientation maps. (1986).
draining veins, even those smaller than Second, their choice of threshold, a criti- 7. Frostig R. D., Lieke, E., Tso, D. Y. & Grinvald, A.
100 m (cortical vasculature image, cal step in obtaining single-condition Proc. Natl. Acad. Sci. USA 87, 60826086
(1990).
Fig. 2a; vascular artifacts Fig. 2c and gi). maps, was arbitrary. A cocktail blank
Such large signals may spread 37 mm (average response to four stimuli, a rough 8. Kim, D.-S., Duong, T. Q. & Kim, S.-G. Nat.
Neurosci. 3, 164169 (2000).
from the sites of electrical activity (A. approximation for uniform cortical acti-
9. Frahm, J. Merboldt, K. D., Hanike, W.,
Shmuel et al., ISMRM abstract, in press; vation elicited by many stimuli) was used Kleinschmisdt, A. & Boeker, H. NMR Biomed.
D. Shoham and A.G., unpublished instead of a real blank (no stimulus). 7, 4553 (1994).
results). Most important, signals from Third, one would expect a small negative 10. Malonek, D. & Grinvald, A. Science 272,
such draining veins may be stimulus-spe- BOLD signal also for the orthogonal ori- 551554 (1996).
cific and highly reproducible. Therefore, entation, rather than a flat line or a small 11. Vanzetta, I. & Grinvald, A. Science 286,
use of differential imaging, high thresh- positive signal (Fig. 2e and f in ref. 8). 15551558 (1999).
olds, signal averaging and statistics does This last reservation may suggest that cur- 12. Logothetis, N. K., Guggenberger, H., Peled, S.
not guarantee removal of such artifacts. rent signal-to-noise ratio is not good & Pauls, J. Nat. Neurosci. 2, 555562 (1999).
Single-condition mapping, in which the enough yet. Such single-condition map- 13. Hu, X., Le, T. H & Ugurbil, K. Magn. Reson.
Med. 37, 877884 (1997).
response to a single stimulus is imaged ping is difficult even with optical imag-
directly, thus has great advantages over ing. (Cocktail blanks are commonly used 14. Engel, S. A., Glover, G. H. & Wandell, B. A.
Cereb. Cortex 7, 181192 (1997).
differential imaging and should be the in optical imaging studies as well 3 .)
15. Menon, R. S., Ogawa, S., Strupp, J. P. &
ultimate goal of high-resolution func- Therefore, future work should emphasize Ugurbil, K. J. Neurophysiol. 77, 27802787
tional brain imaging. increasing signal-to-noise ratio, and addi- (1997).

nature neuroscience volume 3 no 2 february 2000 107


2000 Nature America Inc. http://neurosci.nature.com

brief communications

Rapid, synaptically driven provide a flexible and informationally rich engram for cerebel-
lar motor learning.
increases in the intrinsic Long-term synaptic potentiation and depression (LTP and
LTD) are considered computationally powerful, in part because
excitability of cerebellar they allow for the synapse-specific modification of a large array
of inputs. Less attention has been paid to the idea that information
deep nuclear neurons storage may involve activity-dependent changes in the intrinsic
excitability of neurons. This type of plasticity would affect post-
synaptic signals evoked by many (or possibly all) synapses imping-
Carlos D. Aizenman and David J. Linden ing on a neuron, depending on the identity and location of the
intrinsic conductances altered, thereby creating a more global
Department of Neuroscience, Johns Hopkins University School of Medicine,
725 N. Wolfe Street, Baltimore, Maryland 21205, USA change in signal integration. Slow, activity-dependent changes in
the intrinsic excitability of both invertebrate and mammalian neu-
Correspondence should be addressed to D.L. (dlinden@jhmi.edu)
rons have been reported in cell culture systems1,2. Persistent
The neurons of the cerebellar deep nuclei are implicated in certain changes in intrinsic neuronal excitability can also be induced
forms of motor learning such as associative eyeblink condition- somewhat more rapidly. In the mollusk Hermissenda, associative
ing, partly because increases in their firing rates parallel acquisi- pairing of light and rotation produces a long-term increase in the
tion of the conditioned response. Here we demonstrate that these intrinsic excitability of type B photoreceptors due to a reduction
2000 Nature America Inc. http://neurosci.nature.com

neurons can show persistent increases in their intrinsic excitabil- of K+ currents3. A similar effect is observed in CA1 pyramidal
ity following a Ca 2+ load imposed by synaptic activation of neurons of the rabbit hippocampus following trace eyeblink con-
NMDA receptors or direct current injection. This phenomenon, ditioning4. In hippocampal LTP, increases in the probability of a
together with use-dependent alterations in synaptic strength, may cell firing an action potential for a given size EPSP accompanies

a b
Fig. 1. Tetanization of excitatory synapses
produces a sustained increase in intrinsic
excitability of DCN neurons (a) Averaged
time course of the number of spikes evoked
during 200 ms, depolarizing current test
pulses. Control experiments (open circles)
represent the baseline. Following a synaptic-
conditioning tetanus (at arrowheads), num-
ber of spikes evoked by the test stimulus
slowly increases (filled circles). (b, c)
Representative traces evoked by a +0.5 nA
test pulse in tetanized and control experi-
ments at the time points indicated by aster- d
isks. Traces evoked by subthreshold test
pulses (0.1 and +0.1 nA; b) demonstrate
that Rinput remains stable when the spiking
evoked by a larger depolarizing test pulse
increases. Examples are shown from two c
different cells, one that fired in regular spik-
ing mode (top traces) and one that fired in
burst mode (bottom traces). (d) A cumula-
tive probability distribution shows the
resulting increase in the number of spikes
measured at 2025 min for tetanized and e
control groups shown in (a). (e) The num-
ber of spikes evoked by depolarizing test
pulses is plotted as a function of the ampli-
tude of injected current for four separate
experiments. Solid lines represent the func-
tion immediately before and dashed lines 20
min after the synaptic tetanus. Error bars for
(a) and (e) represent s.e. Experiments used
coronal slices of 1315 day-old rat cerebel-
lum as described9. Slices recorded using
microelectrodes (80150 M) containing 3
M potassium acetate were maintained at
33C in an interface chamber and perfused
with ACSF containing 126 mM NaCl, 5 mM
KCl, 3 mM CaCl2, 1 mM MgSO4, 26 mM
NaHCO3, 1.25 mM NaH2PO4, 20 mM
D-glucose and 0.2 mM picrotoxin and bub-
bled with 95% O2/5% CO2.

nature neuroscience volume 3 no 2 february 2000 109


2000 Nature America Inc. http://neurosci.nature.com

brief communications

a tion storage for associative eye-blink condi-


tioning 8. Microelectrode recordings were
made from neurons in the DCN of juvenile
rat cerebellar slices as described9,10. Picrotox-
in (200 M) was added to block GABA A
receptors. To record changes in intrinsic
excitability, the cell was tonically hyperpolar-
ized to suppress spontaneous spiking, and a
b short depolarizing test pulse (250 ms, 0.10.5
nA), which evoked a small number of action
potentials (typically 14), was then used as a
baseline measure. Repeated application of this
test pulse alone did not alter the number of
evoked spikes, as stable responses could be
recorded for more than > 25 min (Fig. 1a and
c). After a baseline period, an excitatory
synaptic tetanus was applied to the DCN neu-
c ron via a stimulating electrode placed in the
2000 Nature America Inc. http://neurosci.nature.com

adjacent white matter. The tetanus (10 high-


frequency bursts applied at 4 Hz; each burst
comprising 10 pulses delivered at 100 Hz) was
repeated 5 times. In 8 of 9 cells, the number
of spikes evoked by the test pulse increased in
response to this synaptic-conditioning tetanus,
stabilizing in 1520 minutes (Fig. 1a and b).
The number of spikes evoked by the test pulse
d was significantly increased by the tetanus
(n = 9; before, 4 1; increase, 8 2; p < 0.01,
Students t-test; Fig. 1d).When Rinput was mea-
sured using a hyperpolarizing current pulse,
no significant change was observed 20 min
after the tetanus (114 10% of baseline,
n = 4). Furthermore, synaptic tetanization did
not produce a significant change in the ampli-
tude of the first spike evoked by the depolar-
izing test pulse (98 2% of baseline, n = 9).
Experiments were excluded if the amplitude
of the first evoked spike decreased by >10%
Fig. 2. Sustained increases in intrinsic excitability of DCN cells require Ca2+ influx. (a) Averaged at any point in the recording, if the cell exhib-
time course and sample traces from experiments in which 50 M D-AP5 was applied before ited spontaneous spiking between test stim-
synaptic tetanization. (b) Sustained increases in spike firing could also be induced by applying an uli or if the recording could not be
intracellular conditioning tetanus consisting of depolarizing current pulses (indicated by arrow-
maintained for 20 min following the condi-
head; see text). (c) Bath-applied 200 M Cd2+ blocked increases in spiking induced by the same
intracellular conditioning stimulation given in (b). (d) Changes in the late/peak EPSP amplitude
tioning stimulus. Inputoutput curves were
ratio, measured 1520 minutes after synaptic tetanization under different experimental condi- then plotted relating injected depolarizing
tions. There was no significant change in the amplitude of the EPSP peak (left). Arrows indicate current to the number of evoked spikes
EPSP waveforms sampled 20 minutes post-tetanus. immediately before or 20 minutes after the
synaptic conditioning tetanus (Fig. 1e).
Increased excitability was clearly associated
with reduced spike threshold in three of four
increases in synaptic strength. This phenomenon has been called cells. Additionally, in 2/4 cells, maximum spiking rate evoked by
EPSP-to-spike (ES) potentiation. However, as ES potentiation the largest test pulses increased. These results indicate that exci-
is generally occluded by GABAA receptor antagonists, it is pro- tatory synaptic tetanization produced a change in intrinsic post-
posed not to reflect changes in intrinsic postsynaptic conduc- synaptic excitability that did not merely reflect alteration in the
tances, but rather to represent a decrease in the ratio of microelectrode or the general health of the impaled cell.
feedforward inhibition to excitation yielding a net increase in We wished to determine the initial signals required for this
excitability5. However, based on indirect measures using field change in intrinsic excitability. The synaptic conditioning tetanus
potentials6 or intracellular recording with direct current injection evoked spiking (66 12 spikes per burst, n = 9, range, 17130
(but without blocking GABA receptors, allowing confounding spikes per burst), but the number of spikes evoked during the
changes in tonic inhibitory drive)7, others claim that ES poten- synaptic conditioning tetanus was uncorrelated with resulting
tiation reflects an increase in intrinsic excitability. changes in excitability (increase in number of spikes evoked by
The neurons of the deep cerebellar nuclei (DCN) integrate a test pulse; r = 0.34, p > 0.10). To further characterize the
excitatory and inhibitory inputs representing several streams of requirements for this effect, synaptic conditioning tetani were
sensory/motor information, and are a proposed site of informa- applied during bath application of 50 M D-AP5, an NMDA

110 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

brief communications

receptor antagonist. The number of spikes induced during the val) with the resulting increase in the number of spikes. We saw
synaptic conditioning tetanus in D-AP5 was adjusted so that it no correlation between these two measures (r = 0.1, p > 0.1),
was not significantly different from that without drug (p = 0.498, indicating that the cells firing mode did not correlate with its
Kolmogorov-Smirnov). With D-AP5, the conditioning tetanus ability to undergo long-term changes in intrinsic excitability.
failed to produce an increase in the number of spikes evoked by Moreover, we did not observe a strong tendency for cells to switch
the test pulse (0.4 1 spikes, p = 0.83, Students t-test, n = 5; firing mode following the conditioning.
Fig. 2a), indicating that Ca2+ influx through NMDARs is required. Our main finding is that either synaptically driven activation
However, this requirement for NMDAR activation does not pre- of NMDA receptors or Ca2+ influx driven by direct depolarization
clude contribution of other receptors such as mGluRs or AChRs can trigger a rapid and persistent increase in the intrinsic excitabil-
to this effect. ity of DCN neurons. This increase took the form of a reduction
Depolarizing pulses that evoke substantial spiking in DCN in spike threshold and/or an increase in the maximum firing rate.
neurons cause large intracellular Ca2+ transients in both the soma At present, we have no direct evidence to implicate any particular
and dendritic arbor9,12, sufficient to evoke LTP of IPSPs9. We then conductance in this effect. However, the excitability of these cells is
tested whether depolarizing current pulses alone could affect the strongly modulated by a hyperpolarization-activated cation current
excitability of DCN neurons without synaptic stimulation. An (Ih), a low threshold voltage-sensitive Ca2+ current (ICa(T))and an
intracellular conditioning stimulus (pulse amplitude, +0.5 nA; apamin-sensitive Ca2+-gated K+ current10. As the increase in intrin-
duration, 100 ms; applied at 4 Hz and delivered 5 times through sic excitability seemed to require a Ca2+ transient, it is possible that
the recording electrode) caused a marked, gradual increase in the the relevant ion channels were altered by a Ca2+-dependent sig-
2000 Nature America Inc. http://neurosci.nature.com

number of spikes evoked by the test pulse (increase of 6 2 naling cascade such as PKC or Ca2+-sensitive adenylyl cyclase/PKA.
spikes; p = 0.016, Students t-test, n = 7, Fig. 2b). When influx of Activation of these signaling systems produces transient effects on
Ca2+ was prevented by 200 M Cd2+, spiking did not significant- intrinsic excitability in hippocampal neurons, at least in part,
ly increase (1 0.3; p = 0.097, Students t-test; n = 4, Fig. 2c). through actions on Ca2+-gated K+ channels14,15.
Alterations in dendritic or somatic voltage-gated channels How might alterations of intrinsic excitability of DCN neu-
could alter the kinetics of EPSPs. To test this, we delivered a rons contribute to information storage in the cerebellum?
synaptic conditioning tetanus after collecting a 10-min baseline Although not directly tested, the Ca2+ transient produced by a
of EPSPs. The synaptic tetanus caused an inconsistent change in burst-and-pause stimulus delivered to the GABAergic Purkinje
the peak EPSP amplitude ranging from LTP to LTD with many cellDCN synapses9 probably would also be a sufficient signal to
cases of no change at all. To analyze EPSP kinetics, amplitude induce this change. This could provide a framework for het-
measurements were taken at both the peak and a time at which erosynaptic interaction between excitatory and inhibitory inputs
the averaged baseline EPSP waveform had decayed to 50% of its to the DCN. In addition, a striking observation regarding asso-
peak amplitude. These measurements were then normalized to ciative eyeblink conditioning is that extracellular records of spike
the peak amplitude of each EPSP and analyzed as the ratio of firing in the DCN show patterns of increased activity that cor-
late/peak EPSP amplitude. Synaptic conditioning significantly relate with the development of the conditioned response8. The
increased this ratio (121 8% of baseline, p = 0.026 Students t- most common explanation for this observation is that these
test, n = 13) even when there was no change in the peak ampli- increases reflect decreased inhibitory drive from Purkinje cells
tude, indicating that conditioning caused the EPSP to decay more and/or increased excitatory drive from mossy fibers. Our results
slowly (Fig. 2d). As in experiments in Fig. 1b and c, Rinput was suggest that increases in intrinsic excitability could potentially
unaltered by the synaptic tetanus (106 3% of baseline, n = 13, complement input-specific alterations in synaptic strength to
Fig. 2d). EPSP broadening could be partially suppressed by give rise to a flexible and informationally rich engram.
50 M D-AP5 (109 2, p = 0.02 Students t-test, n = 4). As a test
of whether this broadening could result from increases in the ACKNOWLEDGEMENTS
proportion of slower NMDA to faster AMPA currents underly- We thank C. Hansel, A. Kirkwood, H.-K. Lee and S. Morris for suggestions. This
ing the EPSP, the experiment was repeated with an AMPA recep- work was supported by MH01590 and the Develbiss Fund (D.J.L.) and a
tor antagonist, NBQX (10 M). A substantial EPSP could be fellowship from the Howard Hughes Medical Institute (C.D.A.).
evoked in the presence of NBQX even when the cell was held at
negative membrane potentials (below 80 mV), probably because RECEIVED 25 OCTOBER; ACCEPTED 16 DECEMBER 1999
of Mg2+-insensitive NR2D receptor subunits found in the DCN11.
This NMDAR-mediated EPSP also showed a broadening of the 1. Turrigiano, G., Abbot, L. F. & Marder, E. Science 264, 974977 (1994).
EPSP waveform (116 7%, p = 0.03 Students t-test, n = 12), 2. Desai, N. S., Rutherford, L. C. & Turrigiano, G. Nat. Neurosci. 2, 515520
(1999).
suggesting that the observed change in EPSP decay kinetics 3. Alkon, D. L. J. Exp. Biol. 112, 95112 (1984).
involved either a change in intrinsic postsynaptic conductances, 4. de Jonge, M. C., Black, J., Deyo, R. A. & Disterhoft, J. F. Exp. Brain Res. 80,
as observed in hippocampal CA1 pyramidal neurons13, or, alter- 456462 (1990).
5. Abraham, W. C., Gustafsson, B. & Wigstrm, H. J. Physiol. (Lond.) 394,
natively, a selective change in the NMDAR kinetics. 367380 (1987).
We noted a high degree of heterogeneity in the firing modes of 6. Jester, J. M, Campbell, L. W. & Sejnowski, T. J. J. Physiol. (Lond.) 484,
the cells sampled for these experiments, which ranged in a con- 689705 (1995).
7. Pugliese, A. M., Ballerini, L., Passani, M. B. & Corradetti, R. Neuroscience 62,
tinuum from regular spiking (Fig. 1b, top traces) to burst firing 10211032 (1994).
(Fig. 1b, bottom traces). This heterogeneity may reflect different 8. Raymond J. L., Lisberger S. G. & Mauk M. D. Science 272,11261131 (1996).
modulatory states of the DCN neurons10. We tested whether the 9. Aizenman, C. D., Manis, P. B. & Linden, D. J. Neuron 21, 827835 (1998).
10. Aizenman, C. D. & Linden, D. J. J. Neurophysiol. 82, 16971709 (1999).
propensity of the cell to burst correlated with the amount of 11. Cull-Candy, S. G. et al. Neuropharmacology 37, 13691380 (1998).
change in intrinsic excitability resulting from synaptic or depo- 12. Muri, R. & Knpfel, T. J. Neurophysiol. 71, 420428 (1994).
larization conditioning by relating the averaged first inter-spike 13. Hess, G. F. & Gustafsson, B. Neuroscience 37, 6169 (1990).
14. Baraban, J. M., Snyder, S. H. & Alger, B. E. Proc. Natl. Acad. Sci. USA 82,
interval during a test pulse within the baseline period (cells with 25382542 (1985).
a greater propensity to burst have a shorter first interspike inter- 15. Madison, D. V. & Nicoll, R. A. J. Physiol. (Lond.) 372, 221244 (1986).

nature neuroscience volume 3 no 2 february 2000 111


2000 Nature America Inc. http://neurosci.nature.com

articles

A metabotropic glutamate
receptor variant functions as a
taste receptor
Nirupa Chaudhari1, Ana Marie Landin and Stephen D. Roper1

Dept. of Physiology and Biophysics and 1Program in Neuroscience, University of Miami School of Medicine, P.O. Box 016430 (R430), Miami, Florida 33101, USA
Correspondence should be addressed to N.C. (nchaudhari@miami.edu)

Sensory transduction for many taste stimuli such as sugars, some bitter compounds and amino acids
is thought to be mediated via G protein-coupled receptors (GPCRs), although no such receptors that
2000 Nature America Inc. http://neurosci.nature.com

respond to taste stimuli are yet identified. Monosodium L-glutamate (L-MSG), a natural component
of many foods, is an important gustatory stimulus believed to signal dietary protein. We describe a
GPCR cloned from rat taste buds and functionally expressed in CHO cells. The receptor couples neg-
atively to a cAMP cascade and shows an unusual concentrationresponse relationship. The similarity
of its properties to MSG taste suggests that this receptor is a taste receptor for glutamate.

Chemoreceptor cells in taste buds monitor the chemical envi- implicates a metabotropic receptor similar or identical to mGluR4
ronment in the oral cavity and generate signals that lead to taste in taste transduction for L-glutamate20. Such evidence includes the
perceptions. Taste transduction for simple salts involves altered findings that mGluR4 is expressed in rat taste buds17,21 and that
permeation of the receptor cell membrane by ions such as Na+, K+ an mGluR4-selective ligand, L-AP4, mimics the taste of L-MSG in
or H+ (ref. 1). The resulting receptor currents in taste bud cells conditioned taste aversion in rats17 and in human psychophysical
stimulate neurotransmitter release to excite sensory afferents, measurements22. Further, both L-MSG and L-AP4 interact syner-
ultimately leading to perceptions such as salty or sour. Taste gistically with nucleotide monophosphates to elicit umami taste
transduction for larger organic molecules such as sugars, amino responses23,24. Additionally, stimulating taste buds with glutamate
acids or a heterogeneous collection of compounds that elicit per- decreases cellular cAMP (X. Zhou & N. Chaudhari, Chem. Senses
ception of bitterness, is thought to include binding at specific 22, 834, 1997) and alters membrane conductances25, a signaling
receptors on the taste cell plasma membrane1,2. cascade also triggered by mGluR4. Collectively, these findings are
Some of the proteins that orchestrate this plethora of sensory consistent with the transduction of L -glutamate taste by an
transduction mechanisms have been identified using molecular mGluR4-like receptor. Nevertheless, several lines of evidence indi-
biological methods. Taste receptor cells express G proteins, includ- cate apparent discrepancies between umami taste and the proper-
ing -gustducin3, -transducin4, a number of additional G sub- ties of mGluR4. The concentrations of glutamate needed to elicit
units 5,6, several G subunits and a taste-specific G 7. taste and to activate the neurotransmitter receptor mGluR4 differ
Phosphodiesterases4 and a cyclic nucleotide-gated channel8 cloned markedly. The detection threshold for L-MSG in recordings from
from mammalian taste buds could potentially participate in sen- sensory afferents is 0.10.3 mM in juvenile and 13 mM in adult
sory transduction pathways. Epithelial sodium channels demon- rodents26,27, whereas mGluR4 requires glutamate in the micro-
strated in taste buds presumably underlie salty and sour molar range. Further, the ability of glutamate agonists to stimu-
transduction911. Detectable receptor activity for bitter stimuli late mGluR4 does not correlate fully with their umami taste28.
is found in membrane preparations from taste tissue12. Although Additionally, umami taste does not seem to be blocked by a known
a number of candidate taste-GPCRs have been proposed1315, their antagonist of mGluR423. These observations suggest that the recep-
functional significance in taste transduction has not been estab- tor(s) transducing umami taste may differ significantly from
lished2. This report describes the cloning and functional charac- mGluR4, particularly in the glutamate-binding domain.
terization of a taste receptor and its natural taste stimulus. The glutamate-binding domain of the mGluR is contained
Sweet, sour, salty, bitter and (arguably) umami constitute basic within the large extracellular N terminus. Although detailed
taste qualities. Umami denotes the taste of the glutamate moiety structural information is lacking, a model of the N terminus of
in monosodium L-glutamate (L-MSG), a compound that occurs mGluRs is based on the structure of a bacterial periplasmic
naturally in protein-rich and other foods. Taste transduction for leucine-isoleucine-valine binding protein (LIVBP)29. Experi-
glutamate is hypothesized to entail stimulation of neurotransmit- mental verification of this model includes mutation of contacting
ter-like ionotropic and metabotropic glutamate receptors1618. A amino acids29, expression of truncated extracellular domains that
number of ionotropic glutamate receptors are expressed in lingual retain binding characteristics30 and chimeric receptors with dis-
tissue, although none seems preferentially localized to taste buds17. tinct agonist sensitivities31. Thus, the extracellular N terminus
Metabotropic glutamate receptors (mGluR18) constitute a fam- might be a plausible site for differences between neurotransmit-
ily of GPCRs that are found in many neuronal cells19. In taste recep- ter and taste receptors for glutamate. We found an unusual vari-
tor cells, molecular, physiological and behavioral evidence ant of mGluR4, taste-mGluR4, expressed in lingual epithelium.

nature neuroscience volume 3 no 2 february 2000 113


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 1. The 5 end of mGluR4


cDNA from taste papillae a
contains novel sequence
derived from an intron.
(a) The 5 end of mGluR4
cDNA from taste papillae
(lower lines) is aligned with
the corresponding region of
mGluR4 cDNA from brain
(upper lines). Amino acid
(bold) and translated
nucleotides (regular) for each
sequence are numbered. A
stop codon (), in frame with
the long open reading frame, b
is found within the novel
region of the cDNA from
taste tissue. Sequence identity
between the two cDNAs
begins abruptly at the codon
2000 Nature America Inc. http://neurosci.nature.com

for R291. This position also


corresponds to the location of
intron a (). (b) Schematic
showing the full-length cDNA
for mGluR4 from brain,
including 5 and 3 untrans-
lated regions (line), translated
region (shaded bar) and
regions encoding seven puta-
tive transmembrane segments
(gray stripes). The 800-bp seg-
ment (double line) expressed c d
in rat taste buds17 as well as
the 3 and 5 RACE products
(**) we characterized are
shown above the brain-
derived mGluR4 cDNA. The
corresponding genomic orga-
nization shows two interven-
ing sequences. The last 60 bp
at the 3 end of intron a (in
black) are identical to the first
60 bp at the 5 end of the
taste-derived cDNA; the taste-derived cDNA includes part of intron a followed by exon N1. (c) mGluR4 mRNA from brain includes several exons
upstream of N0, whereas mGluR4 mRNA derived from taste tissue begins with an extended exon N1 spliced to exon N2. In the taste-derived mRNA, the
presence of the upstream stop codon prevents translation of the first 128 bp (white); translation presumably initiates at the next methionine codon (M309
in brain-derived cDNA). (d) Predicted transmembrane topology of brain- and taste-mGluR4 showing the truncated extracellular N terminal domain fol-
lowed by seven putative transmembrane helices and cytoplasmic C terminus. The LIVBP-like glutamate-binding domain29 and its truncated version are
shown as heavy lines.

The corresponding protein is predicted to lack approximately lium17,21. However, it is unclear whether mGluR4 in taste buds is
half the N terminus, including a large portion of the LIVBP-like identical to that in the brain. Earlier RT-PCR and in-situ hybridiza-
putative glutamate-binding domain. Taste-mGluR4 cDNA tion analyses focused on an ~800-bp core region conserved among
expressed in CHO cells conferred sensitivity to L-glutamate at mGluR1-8 (Double line in Fig. 1b). The N terminus of mGluRs
concentrations ~100-fold higher than needed for brain-mGluR4, comprises a large extracellular glutamate-binding domain29. The
and its expression level was negatively coupled to cAMP con- cytoplasmic C terminus of mGluRs participates in interactions
centration. These findings correspond well with the concentra- with G proteins. Because N and C termini of mGluRs determine
tions of glutamate needed to elicit umami taste and resolve the essential functional characteristics, we analyzed the corresponding
discrepancy between neurotransmitter receptors for glutamate sequences for mGluR4 expressed in taste tissue.
and taste receptors for glutamate. Some of these results were pre-
sented in abstracts (A. Fedorov & Chaudhari, N., Chem. Senses Full-length cDNA for mGluR4 from taste tissue
23, 593, 1998; A. M. Landin et al., Chem. Senses 24, 586, 1999). In the brain, mGluR4 mRNA is found in two forms, mGluR4a and
mGluR4b, which differ at the 3 end32. The longer mRNA includes an
RESULTS exon containing an in-frame stop codon, and thus generates a short-
In-situ hybridization shows that mGluR4 is expressed in rat taste er protein product, mGluR4a. To analyze the C terminus of mGluR4
buds and cannot be detected in surrounding non-sensory epithe- in taste cells, we used poly(A)RNA from taste (circumvallate and

114 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

foliate) papillae and gene-specific a b


primers located in the previously
characterized 800-bp core region.
We performed 3 RACE (rapid
amplification of cDNA ends)
reactions and cloned the longest
specifically amplified bands.
DNA-sequence analysis of repre-
sentative clones yielded 100%
identity with mGluR4a cDNA
from the brain33. We also carried
out RT-PCR with a primer pair
straddling the alternatively spliced
exon. The principal amplification
product was confirmed by DNA
sequence analysis to be mGluRa.
A faint band corresponding in
size to mGluR4b was detected Fig. 2. The truncated mRNA in taste papillae is a mature mRNA. (a) RT-PCR with a reverse primer (R) in exon
2000 Nature America Inc. http://neurosci.nature.com

only in occasional PCRs (not N2 and forward primers B (within exon N0) or T1, T2, T3 (within intron a) using poly(A)RNA from taste tissue
shown). Thus, we conclude that or from brain. With taste RNA, PCR product is visible in lanes amplified with T1-R and B-R primer pairs but not
the C-terminal sequence in taste with the T2-R or T3-R primer pairs, demonstrating that only a portion of intron a sequence is present in the
tissue was predominantly of the RNA. With brain RNA used as template, PCR products are clearly seen with all three primer pairs, T1-R, T2-R
mGluR4a type. and T3-R, implying that sequences from intron a are present in unspliced precursor for the known mGluR4
Gene-specific reverse primers mRNA. A 1-kb ladder (BRL) is in the far left lane. Schematics on the right show RNA templates postulated to
yield the PCR products obtained (taste-mGluR4, precursor of brain-mGluR4 and mature brain-mGluR4,
in the 800 bp core region were
respectively). (b) RNase protection assay demonstrates that taste tissue contains significant amounts of the
also used in 5 RACE reactions. truncated mGluR4 RNA. [32P]-labeled probe was hybridized with poly(A)RNA from liver, cerebellum (0.05 g,
Previously cloned cDNAs for rat cer) and taste papillae (23 g) from adult (ad) or juvenile (juv) rats. Lengths are indicated in nucleotides; RNA
mGluR4a include a 5 untrans- markers are on the left, and RNase-protected fragments are on the right. A schematic of the probe used (right)
lated region (5 UTR) of either shows exons (gray) and the segment of intron a found in taste-derived mGluR4 cDNA (black).
69 bp 33 or 854 bp 29. Thus, we
estimated that our 5 RACE
products should range between
1450 and 2235 bp. Using brain poly(A)RNA to validate the (precursor) RNA rather than a mature mRNA. Because introns
method, amplification products extending to 2000 bp were are spliced out intact, a precursor RNA should include the com-
obtained as expected. In contrast, the 5 RACE product obtained in plete sequence of intron a. This prediction was tested by RT-PCR
parallel from taste tissue terminated abruptly at approximately 600 (Fig. 2a). We used three forward primers (T1, T2 and T3) along
bp. Similarly truncated products from taste-derived mRNA result- intron a. The reverse primer (R) was selected from a separate
ed from 5 RACE reactions with at least four different gene-spe- downstream exon to preclude amplification from genomic DNA.
cific reverse primers and two sources of reverse transcriptase. From brain poly(A)RNA, approximately equal amounts of prod-
uct were detected for all three reactions using forward primers
A distinct mGluR4 cDNA found in taste tissue within intron a (Fig. 2a). This result implies that intron a may be
Sequence analysis of the cloned RACE product indicated that the a late-spliced intron and that brain poly(A)RNA contains pre-
5 terminal 4060 bp of the taste-derived cDNA clones were not cursor RNAs that include the complete intron. In contrast, taste
similar to any region of brain mGluR4 cDNA (Fig. 1a). A stop poly(A)RNA showed amplification product only from the far-
codon was found near the 5 end, in-frame with the long open thest-downstream intronic primer, T1, and not from upstream
reading frame, implying that the 5 end is untranslated sequence. primers T2 or T3. Thus, poly(A)RNA in taste tissue included only
We found two potential start codons in frame, 102 bp and 189 bp a short segment from the 3 end of intron a, suggesting that the
downstream of the stop codon. Thus, the sequence of mGluR4 truncated mGluR4 cDNAs obtained in 5 RACE were probably
cDNA from taste tissue is unique for the first ~60 bp and then is derived not from an unspliced precursor RNA, but from a mature
identical for at least 2220 bp to mGluR4a cDNA from brain. mRNA. A forward primer (B) located in the next exon upstream
The point of divergence between unique and identical (N0), amplified from the previously known mGluR4 mature
sequences for taste- and brain-derived cDNAs, located at amino mRNA, served as a control. PCR product from mGluR4 mRNA
acid R291 of mGluR4a, resembles a splice acceptor site. To test lacking this intron was detected in poly(A)RNA from both brain
this, we analyzed a genomic fragment amplified from this region and taste papillae. Precursor RNAs are typically found in tissue
and determined that an intron (intron a, Fig. 1b) interrupts the at considerably lower concentration than their respective mature
codon for R291. The 3 end of intron a is identical to the ~60-bp mRNAs. In taste tissue, the low concentration of mature full-
unique sequence at the 5 end of the taste-derived mGluR4 length mGluR4 mRNA17 precludes detecting its precursor RNA
cDNAs (Fig. 1a and b). One additional intron was also identified (as RT-PCR products with T3 and T2 primers).
further downstream. RNA secondary structure can cause premature termination
of reverse transcripts and yield truncated products in 5 RACE.
Taste-tissue mGluR4 is a mature mRNA This did not seem to be the case for the truncated taste-mGluR4
The presence of intron sequence in the taste-derived mGluR4 cDNA because brain poly(A)RNA did yield long 5 RACE prod-
cDNA raised the possibility that it was amplified from a nuclear ucts, and because precursor RNAs in brain samples were readily

nature neuroscience volume 3 no 2 february 2000 115


2000 Nature America Inc. http://neurosci.nature.com

articles

band, 254263 nucleotides in


a b length, as might be expected for an
RNA that included the 178
nucleotides of exon (N 1 plus N 2)
sequence and extended 7685
nucleotides forward into intron a.
This protected band was consistent
with the mGluR4 cDNA cloned in
the 5 RACE experiments. The band
was not detected in cerebellar RNA
lanes. Thus, we designate this form
of mGluR4 RNA, which includes a
part of intron a, as taste-mGluR4.
We note that the ~178-bp band
Fig. 3. Taste-mGluR4 is activated by glutamate at much higher concentrations than brain-mGluR4. derived from brain-mGluR4 is
(a) Immunoblot of CHO cells stably transfected with brain-mGluR4 (B) or taste-mGluR4 (T) shows a promi- sharp, as expected, whereas the
nent immunoreactive band of predicted molecular weight (~102 kDa and 68 kDa, respectively) when probed broad taste-mGluR4-derived band
with an antibody against the shared C terminus of both forms. CHO cells transfected with non-recombinant indicates more heterogeneity in
2000 Nature America Inc. http://neurosci.nature.com

vector (con) do not show either band. Extracts of cerebellum (cer) and brain-mGluR4 expressing CHO cells length. The 5 ends of mRNAs fre-
also show a higher band, presumably a dimerized receptor at 208 kDa. (b) CHO cells expressing brain- quently show such heterogeneity,
mGluR4 () and taste-mGluR4 () both respond to glutamate by decreasing cAMP levels. Cells were stimu-
spanning 315 nucleotides.
lated with L-glutamate in the presence of forskolin and IBMX (see Methods). Six independent experiments
were carried out, each including triplicate wells of cells for each concentration. Three of the experiments
We observed the broad band
were conducted on lines of clonal transformants and another three on non-clonal lines of stably expressing corresponding to taste-mGluR4
CHO cells. The values represent mean s.e.; the curves are a sigmoidal fit to the data. Control cells, trans- mRNA in three separate protection
fected with the non-recombinant vector did not significantly alter cAMP levels when stimulated with L-gluta- experiments using different batch-
mate (1 mM, 127 16.0%; 10 mM, 132 19.0%; n = 9). The absolute values of cAMP per well in es of poly(A)RNA from taste tissue
forskolin-stimulated cells in the absence of glutamate were: 6.07 0.60 pmole, 6.88 0.78 pmole and of juvenile rats. Densitometric
4.33 0.42 pmole for cells transfected with brain-mGluR4, taste-mGluR4 and vector, respectively. analysis from three experiments
indicated that taste-mGluR4
mRNA is present at 70120% of
the concentration of brain-
reverse transcribed and amplified through intron a (Fig. 2a). mGluR4 mRNA in taste tissue from juvenile rats. Interesting-
Thus, based on the 5 RACE and RT-PCR analyses above, we ten- ly, the taste-mGluR4 band was found at lower concentration
tatively concluded that taste tissue may contain two forms of when poly(A)RNA from adult rather than juvenile rats was
mGluR4 mRNAone similar to the known mGluR4a33 and used for protection (Fig. 2b).
another with a truncated 5 end.
Because RT-PCR can detect RNAs that are present in minor Taste-mGluR4 is a functional mRNA
(nonphysiological) quantities in cells, we tested whether the trun- To determine whether taste-mGluR4 is a functional mRNA, we
cated mGluR4 RNA found in taste tissue was present at significant generated full-length clones for both forms of mGluR4 in
concentration using RNase protection (Fig. 2b), an independent pcDNA3.1 vector, transfected them into CHO cells and selected
method not based on amplification. The probe for this assay con- stable transfectants. When probed with an antibody against the C
sisted of the last ~400 nucleotides of intron a, followed by 178 terminus of mGluR4a, immunoblots (Fig. 3a) of cerebellar extracts
nucleotides in two consecutive exons, N1 and (part of) N2. No contained a strong band of the expected molecular weight, 102
bands were generated with RNA from liver, a control tissue that kDa, as previously reported34. A presumed dimer at 210 kDa was
does not express mGluR4, demonstrating the specificity and also detected. Clones of cells stably transfected with brain-mGluR4
RNase sensitivity of the probe. The known full-length form of showed prominent bands of the same size as in cerebellum (Fig. 3a).
mGluR4 mRNA (henceforth designated brain-mGluR4) should In contrast, CHO cells transfected with the taste-mGluR4 construct
protect a band of 178 nucleotides, as it includes no sequence from consistently showed a prominent band of 68 kDa, corresponding
intron a. Indeed, consistent with an identity as brain-mGluR4, to the size predicted from the cDNA sequence. Neither the 102-
a protected band of ~180 nucleotides was detected in kDa nor 68-kDa bands was present in parallel lanes containing
poly(A)RNA from cerebellum and, at a lower concentration, from lysates of CHO cells transfected with non-recombinant pcDNA
taste papillae. In addition, we detected two bands corresponding vector. Thus, the truncated taste-mGluR4 mRNA characterized
to precursor nuclear RNA when cerebellar poly(A)RNA was used from taste tissue was indeed a functional mRNA that was translat-
in RNase protection. Because exons N1 and N2 are not contiguous ed into immunologically recognizable protein.
in the genome, precursor RNA protected fragments ~460
nucleotides long (400 nucleotides of intron a plus exon N1) and Taste-mGluR4 is negatively coupled to cAMP
~120 nucleotides (fragment of exon N2). The absence of these In transfected CHO cells, activation of group III mGluRs leads to a
precursor bands in taste samples indicated that genomic DNA suppression of forskolin-stimulated cAMP synthesis35. CHO cells
(which would protect the same size bands as precursor RNA) was stably expressing brain-mGluR4 responded predictably to L-gluta-
not a significant contaminant in the taste samples. mate (Fig. 3b). The EC50 for this response was 2 M glutamate, con-
In addition to bands derived from the known mGluR4 sistent with earlier reports on mGluR4a (5 M)35. Cells expressing
mRNA and its precursor RNA, hybridization with poly(A)RNA taste-mGluR4 displayed no response to L-glutamate at concentra-
from taste papillae yielded an additional band. This was a broad tions of 30 M or below. The EC50 for L-glutamate for taste-mGluR4

116 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

taste. The cAMP levels in control cells transfected with non-


recombinant vector did not change upon treatment with L-AP4.

DISCUSSION
MGluR4 is a metabotropic glutamate receptor originally char-
acterized from the brain. In situ hybridization analyses have
shown that this receptor is also expressed in taste buds17,21. The
present report demonstrates that mGluR4 in taste tissue is
expressed as a structurally and functionally distinct form that we
have termed taste-mGluR4. Taste-mGluR4 is a truncated ver-
sion of the previously characterized brain receptor, and lacks
50% of the receptors extracellular N terminus. This truncation
is particularly significant, because the N terminus of metabotrop-
ic glutamate receptors is believed to contain the glutamate-bind-
Fig. 4. Taste-mGluR4 is activated by L-AP4 at higher concentrations ing domain29, and changes in this region are likely to influence
than is brain-mGluR4. CHO cells expressing brain-mGluR4 (solid bars) the affinity of the receptor for ligands. Indeed, we report here
and taste-mGluR4 (shaded bars) were stimulated with the indicated that the truncated taste-mGluR4 is much less sensitive to L-glu-
concentrations of L-AP4. Six independent experiments were carried out tamate and L-AP4 than the full-length brain form, implying a
2000 Nature America Inc. http://neurosci.nature.com

(three each on clonal and non-clonal lines of stably transfected cells). reduced affinity for these agonists. Importantly, the reduced sen-
For each concentration, triplicate wells of cells were used in each exper- sitivity of taste-mGluR4 corresponds well with the concentra-
iment. The values represent mean s.e. tions of L-glutamate and L-AP4 needed to elicit a response in
gustatory receptor cells in situ. The data are fully consistent with
the interpretation that the novel taste-mGluR4 is a taste recep-
tor for umami (the taste quality elicited by L-MSG).
was calculated to be 280 M glutamate. We considered the possi- The molecular identification and characterization of taste
bility that a low cell-surface density of taste-mGluR4 might explain receptors has lagged behind research on other sensory receptors,
its low efficacy (high EC50). Thus, we examined two separate lines notably, receptors for vision and olfaction. GPCRs are present
of CHO cells expressing taste-mGluR4. Expression levels of taste- in taste tissue13,14; receptors with sequences related to those of
mGluR4 (as determined by immunoblot) were 100-fold and 2-fold the mGluRs15. Although mRNA and/or protein for such candi-
lower, respectively, than in parallel lines expressing brain-mGluR4. date taste receptors has been demonstrated in lingual tissue, the
Nevertheless, the EC50 values for the two lines expressing taste- lack of functional expression has hampered ligand identification
mGluR4 were very similar, 300 and 250 M, respectively. The con- and validation of their physiological significance. One of the chal-
sistently high EC50 for taste-mGluR4 suggests that low receptor lenges for studying the function of taste receptors is the high con-
density does not explain its low efficacy. Instead, the data suggest centrations of stimuli needed. In the case of sugars, salts, and
that taste-mGluR4 is approximately two orders of magnitude less glutamate, the detection thresholds of gustatory sensory cells in
sensitive to L-glutamate than is brain-mGluR4. Direct measurements nerve recordings or behavioral tests are in the range of a few hun-
of affinity will be necessary to confirm this interpretation. dred micromolar and higher. The low sensitivity of taste recep-
Because the concentration of L-glutamate required for taste- tors, which may result from low affinity for ligands, has
mGluR4 was high, we considered that osmotic or other nonspe- complicated binding assays and functional tests. By utilizing a
cific effects might influence the cAMP response. Thus, we high concentration of glutamate and a selective glutamate recep-
stimulated mGluR4-expressing cells with D-glutamate, an isomer tor ligand (L-AP4), we have been able to characterize the func-
that does not elicit umami taste36. Relative to controls, cells stim- tion of taste-mGluR4 in transfected cells.
ulated with 1 mM D-glutamate yielded cAMP levels of 126 17% The taste-mGluR4 cDNA in this report was cloned from pos-
(brain-mGluR4) or 99 3% (taste-mGluR4). Control CHO cells terior (circumvallate and foliate) taste papillae of juvenile rats.
transfected with non-recombinant vector did not alter cAMP con- The threshold concentration for activating taste-mGluR4 (30 M)
centrations upon treatment with either L- or D-glutamate. Thus, matches well with the threshold (100 M) reported for glutamate
receptor-independent mechanisms do not seem to decrease cAMP. taste responses in the glossopharyngeal nerve of juvenile mice26.
Interestingly, taste nerve thresholds in adult mice and rats, at 210
Taste-mGluR4 responds to L-AP4 mM, are significantly higher26,27. In our studies, we found that the
In rat and human behavioral studies, L-AP4 mimics the taste of mRNA for taste-mGluR4 is expressed at lower concentration in
L-MSG17,22. Thus, we predicted that the taste receptor for L-MSG adult than in juvenile rats17 (Fig. 2b), which may explain the
should also be stimulated by L-AP4. We directly tested whether decreased sensitivity to MSG taste in adult rodents.
taste-mGluR4 meets the criterion of a taste receptor by measur- L-AP4 is a highly effective ligand at brain-mGluR4 (ref. 35).
ing its response to L-AP4 in a concentration range effective for L-AP4 mimics the taste of glutamate in rats17 and is an umami
taste. CHO cells expressing brain-mGluR4 responded to L-AP4 stimulus in humans22. Here we show that L-AP4 also stimulates
by suppressing forskolin-stimulated cAMP production. The taste-mGluR4, at concentrations effective as taste stimuli. MAP4,
response seemed to saturate at all concentrations of L-AP4 above an antagonist of brain-mGluR4, fails to block taste nerve
10 M (Fig. 4), as expected from the published EC50 of 0.51.0 responses to glutamate and L-AP4 in chorda tympani nerve
M35. In cells expressing taste-mGluR4, 10 M L-AP4 was inef- recordings23. Tests of MAP4 on cloned taste-mGluR4 may help
fective, whereas 100 M and 1 mM L-AP4 gave progressively larg- to resolve this seeming paradox. Given the drastically altered
er responses. For rats in a behavioral assay, 100 M L-AP4 is near N terminus of taste-mGluR4, it is impossible to predict its
the detection threshold17. Thus, taste-mGluR4 responds to L-AP4 response to the various glutamate analogs known to activate or
over a concentration range similar to that observed for L-AP4 antagonize brain-mGluR4.

nature neuroscience volume 3 no 2 february 2000 117


2000 Nature America Inc. http://neurosci.nature.com

articles

A distinctive feature of umami is the potentiation of glutamate 5 and 3-RACE and RT-PCR. Initial 5 RACE (rapid amplification of
responses by the nucleotide monophosphates of inosine and cDNA ends) reactions were carried out using superscript reverse tran-
guanosine (IMP and GMP). This synergy is well documented in scriptase followed by terminal deoxynucleotidyl transferase (both from
human psychophysical studies37, animal behavioral experiments24, Gibco-BRL, Gaithersburg, Maryland)44. Subsequently, the Marathon
RACE system (Clontech Laboratories, Palo Alto, California) was
gustatory nerve recordings27 and patch-clamp studies25. Synergis-
employed for generating and cloning RACE products. Poly(A)RNA,
tic interactions are also found between L-AP4 and nucleotides23,24, extracted from vallate and foliate papilla, was used to synthesize double
further underscoring the importance of taste-mGluR4 to umami strand cDNA, which was then ligated to an adapter-primer. Nested gene-
transduction. The site of interaction between glutamate and specific primers were designed in the core region of rat mGluR4 cDNA
nucleotides remains to be defined, but might involve synaptic con- sequence within the 800-bp region known to be expressed in taste buds17.
vergence of separate gustatory sensory cells onto common affer- RACE reactions in both directions were carried out using Klentaq DNA
ent fibers, or converging signaling pathways from separate receptors polymerase (Clontech) or Elongase (BRL) to ensure amplification of long
within the same glutamate-sensing taste bud cell. It is also possible products. Annealing steps were at the highest temperature permitted by
that both glutamate and nucleotides interact with a common recep- respective primers. Amplification proceeded for 2530 cycles to mini-
tor molecule. For instance, ligand-binding studies on bovine taste mize nonspecific products. Amplification products were electrophoresed
and blot hybridized to identify mGluR4-related bands, which were then
membranes suggested an allosteric model for nucleotide effects on
cloned into pGEM-T vector (Promega) and sequenced on an ABI
glutamate taste38. The cloned taste-mGluR4 will allow direct tests Sequencer Model 373A.
of such models of receptor-taste stimulus interactions. The following primers based on mGluR4a cDNA33 (with identifying
Our RT-PCR and RNase protection analyses indicate that both
2000 Nature America Inc. http://neurosci.nature.com

amino acid positions) were used in reverse transcriptase-polymerase


taste-mGluR4 and brain-mGluR4 are expressed in taste tissue (Fig. chain reaction (RT-PCR):
2a and b). Glutamate is implicated as a neurotransmitter for taste B: 5 (D266) CGACAAGATCATCAAACTGCCTAC 3
bud afferent synapses39, and brain-mGluR4 could serve as an autore- R: 5 (F455) GAAGTTGACGTTCCTGATGTACT 3
ceptor at such synapses. We suggest that taste-mGluR4 is likely to To isolate a genomic fragment that contained intron a, genomic DNA
function as an umami receptor, whereas brain-mGluR4 may serve was amplified with primers located in cDNA sequence: forward primer B
(above) and a reverse primer at F307N301. The following primers were
as a neurotransmitter receptor at synapses in or near taste papillae.
based on intron sequence derived from genomic clones, and were used to
Glutamate receptors other than mGluR4 may be expressed in map whether the entire intron a was represented in precursor nuclear
gustatory sensory cells. For example, patch-clamp recordings, Co2+ RNAs (Fig. 2a):
uptake, and Ca2+ imaging on rat and mouse taste buds suggest that T1: 5 (48 bp upstream of R292) CAGCTGGGTAGCCTTACATGTCT 3
glutamate activates both ionotropic and metabotropic glutamate T2: 5 (400 bp upstream of R292) TCTGGAGTAGGATCAGGTGGATG 3
receptors18,25,39,40. However, all these experiments exposed both T3: 5 (2000 bp upstream of R292) AAAGGCTGCTATCTCGTGGACT 3;.
apical and basolateral membranes of taste receptor cells to gluta-
mate. Given that bona-fide taste stimulation reaches only the api- RNase protection assay. The template for mGluR4 probe was constructed
cal membrane of taste receptor cells, it is critical to note that by ligating together a genomic fragment and a cDNA fragment at a shared
behavioral studies and nerve recordings (which stimulate only the AflIII site located 32 bp upstream of the junction between intron a and exon
N1 (Figs. 1b and 2b). The genomic fragment contained the downstream
apical membrane) indicate a minimal role for ionotropic-like glu-
400 bp of intron a (extended for 368 bp upstream of the AflIII site). The
tamate receptors in taste transduction for L-MSG17,2224. Thus, baso- 210 bp cDNA segment of the chimeric probe began at the AflIII site in intron
lateral synapses on gustatory receptor cells may include glutamate a and included two sequential exons, N1 and part of N2. The resulting con-
receptors other than taste-mGluR4 (reviewed in ref. 20). struct in pGEM-T vector was used to transcribe an antisense probe (Fig.
Transcripts of several mGluRs, including mGluR1 and mGluR5, 2b) with at least 238 nt complementary to taste-mGluR4 mRNA, as pre-
are alternatively spliced41,42. For mGluR4, alternative splicing gives dicted from the sequence of the longest 5 RACE clone (see Results). [32P]-
rise to receptors with either long or short C termini32. In spite of labeled antisense RNA was transcribed using T7 RNA polymerase.
considerable structural diversity, most alternative-splicing variants of Hybridization and RNase digestion were performed using the High-Speed
mGluRs show only subtle changes in their functional properties. For Hybridization RPA kit from Ambion (Austin, Texas). Protected fragments
were analyzed on denaturing 6% polyacrylamide-urea gels.
instance, mGluR1c and mGluR1a elicit distinct temporal patterns
of Ca2+ release42. The present case of taste-mGluR4 demonstrates
substantially different function of two receptors derived from the Transfection. Full-length cDNAs for brain-mGluR4 and taste-mGluR4
same gene. We do not presently know the molecular mechanism by (see Results) were reconstructed from cerebellar or taste poly(A)RNA
which the truncated mRNA is produced in taste cells. It is possible respectively, by RT-PCR using Elongase (BRL) for high-fidelity amplifi-
that an alternative splice acceptor site is located within intron a and cation. The brain-mGluR4 insert included 7 nucleotides of 5 untrans-
lated region; the taste-mGluR4 insert included 100 nucleotides of
that our 5 RACE reactions did not progress into additional exons
presumed 5 UTR upstream of the first in-frame start codon. Both cDNA
upstream of the ~60 bp in intron a. Nevertheless, the presence of an inserts terminated at their common stop codon (following I912), yield-
in-frame stop codon within these 60 nucleotides implies that any ing the mGluR4a version of the C terminus33. The inserts were cloned
additional exons would constitute 5 UTR of the mRNA and would into the EcoRI site of pcDNA3.1 vector (Invitrogen).
not affect the sequence of the translated protein. An alternative pos- CHO cells were transfected with brain-mGluR4 and taste-mGluR4 con-
sibility is that the origin for transcription of taste-mGluR4 mRNA is structs and with non-recombinant pcDNA3.1 vector, all in parallel, using
located within intron a. Multiple promoters yielding mRNAs with a cationic lipid, DMRIE-C (Gibco-BRL). Cells with stably integrated plas-
distinct 5 exons occur in the mGluR5 gene43. mid were selected and maintained in 300 g per ml G418 starting 24 h
after transfection. The medium consisted of Dulbeccos Modified Eagle
METHODS Medium (D-MEM) supplemented with 10% fetal bovine serum, 2 mM
Tissues and RNA. All tissues were from Harlan Sprague-Dawley rats. glutamine, 1.7 mM proline and 100 units per ml penicillin-streptomycin35.
Circumvallate and foliate taste papillae were dissected from tongue, and Clones were lifted within two weeks and screened for expression by west-
rapidly frozen on dry ice. Poly(A)mRNA was extracted from tissues by ern blot analysis. A single clone for each form was propagated for func-
direct binding to oligo dT-cellulose (FastTrack II kit, Invitrogen, Carlsbad, tional assays. Stably transfected but non-clonal lines of cells from
California). Unless stated otherwise, taste tissue samples were from juve- independent transfections with both forms of mGluR4 and non-recom-
nile (pre-weaning 1620 day-old) rats. binant vector were also maintained and used in functional assays.

118 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Immunoblot analysis. Anti-peptide anti-serum directed against the C- 16. Faurion, A. Are umami taste receptor sites structurally related to glutamate
terminal 18-amino-acid sequence of mGluR4a was raised in rabbits and CNS receptor sites? Physiol. Behav. 49, 905912 (1991).
17. Chaudhari, N. et al. The taste of monosodium glutamate: Membrane
antibody was purified by affinity chromatography (Zymed Laboratories, receptors in taste buds. J. Neurosci. 16, 38173826 (1996).
San Francisco, California). This epitope is shared between both brain- 18. Hayashi, Y., Zviman, M. M., Brand, J. G., Teeter, J. H. & Restrepo, D.
mGluR4 and taste-mGluR4 described here. Lysates of transfected CHO Measurement of membrane potential and [Ca2+ ]i in cell ensembles:
cells were electrophoresed and tested for expression of brain- and taste- Application to the study of glutamate taste in mice. Biophys. J. 71, 10571070
mGluR4 by immunoblot analysis45 on PVDF membrane. Detection was (1996).
19. Conn, P. J. & Pin, J. P. Pharmacology and functions of metabotropic
with alkaline phosphatase-conjugated secondary antibody and CSPD glutamate receptors. Annu. Rev. Pharmacol. Toxicol. 37, 205237 (1997).
chemiluminescent substrate (both from Tropix, Bedford, Massachusetts). 20. Chaudhari, N. & Roper, S. D. Molecular and physiological evidence for
Bands on autoradiographs were densitometrically quantified as needed. glutamate (umami) taste transduction via a G protein-coupled receptor. Ann.
NY Acad. Sci. 855, 398406 (1998).
21. Yang, H., Wanner, I. B., Roper, S. D. & Chaudhari, N. An optimized method
Functional assay. CHO cells, stably transfected with brain-mGluR4, taste- for in situ hybridization with signal amplification that allows the detection of
mGluR4 or non-recombinant pcDNA3.1, were maintained as sub-con- rare mRNAs. J. Histochem. Cytochem. 47 431446 (1999).
fluent cultures and refed every two days to minimize chronic stimulation 22. Kurihara, K. & Kashiwayanagi, M. Introductory remarks on umami taste.
of expressed receptors by glutamate released from dead cells. Cells were Ann. NY Acad. Sci. 855, 393397 (1998).
plated 20 h before assay in a 96-well microtiter plate at 2 104 cells per 23. Sako, N. & Yamamoto, T. Analyses of taste nerve responses with special
reference to possible receptor mechanisms of umami taste in the rat.
200 l well. Fresh medium was replaced for one hour immediately before Neurosci. Lett. 261, 109112 (1999).
assaying receptor function as described35. Briefly, cells were incubated in 24. Delay, E. R. et al. Taste preference synergism between glutamate receptor
Dulbeccos phosphate buffered saline containing 1 mM IBMX for 20 min ligands and IMP in rats. Chem. Senses (in press).
followed by stimulation for 10 min in 10 M forskolin and 1 mM IBMX, 25. Lin, W. & Kinnamon, S.C. Physiological evidence for ionotropic and
2000 Nature America Inc. http://neurosci.nature.com

with or without agonists. Stimulation buffer was rapidly removed, and metabotropic glutamate receptors in rat taste cells. J. Neurophysiol. 82,
20612069 (1999).
cells were lysed in 200 l 0.25% dodecyltrimethyl-ammonium bromide 26. Ninomiya, Y., Tanikukai, T., Yoshida, S., Funakoshi, M. & Tanimukai, T.
in 50 mM acetate buffer (Amersham, Piscataway, New Jersey). Released Gustatory neural responses in preweanling mice. Physiol. Behav. 49, 913918
cAMP in 20% of each lysate was assayed directly using an Amersham EIA- (1991).
based kit and plotted as mean s.e. of 36 experiments, each performed 27. Yamamoto, T. et al. Electrophysiological and behavioural studies on the taste
of umami substances in the rat. Physiol. Behav. 49, 919925 (1991).
with triplicate wells of cells. 28. Monastyrskaia, K. et al. Effect of the umami peptides on the ligand binding
and function of rat mGlu4a receptor might implicate this receptor in the
ACKNOWLEDGEMENTS monosodium glutamate taste transduction. Br. J. Pharmacol. 128, 10271034
This work was supported by grants from NIH/NIDCD (DC 03013) and from Cultor (1999).
29. OHara, P. J. et al. The ligand-binding domain in metabotropic glutamate
Food Science, Inc. We are also grateful for support from the Umami Manufacturers receptors is related to bacterial periplasmic binding proteins. Neuron 11,
Association of Japan during the early stages of this study. We acknowledge technical 4152 (1993).
assistance from Cynthia Lamp and Helena de Carvalho. 30. Han, G. & Hampson, D. R. Ligand binding to the amino-terminal domain of
the mGluR4 subtype of metabotropic glutamate receptor. J. Biol. Chem. 274,
1000810013 (1999).
RECEIVED 20 SEPTEMBER; ACCEPTED 14 DECEMBER 1999 31. Takahashi, K., Tsuchida, K., Tanabe, Y., Masu, M. & Nakanishi, S. Role of the
large extracellular domain of metabotropic glutamate receptors in agonist
selectivity determination. J. Biol. Chem. 268, 1934119345 (1993).
1. Lindemann, B. Taste reception. Physiol. Rev. 76, 719766 (1996). 32. Thomsen, C. et al. Cloning and characterization of a metabotropic glutamate
2. Lindemann, B. Receptor seeks ligand: on the way to cloning the molecular receptor, mGluR4b. Neuropharmacology 36, 2130 (1997).
receptors for sweet and bitter taste. Nat. Med. 5, 381382 (1999). 33. Tanabe, Y., Masu, M., Ishii, T., Shigemoto, R. & Nakanishi, S. A family of
3. McLaughlin, S. K., McKinnon, P. J. & Margolskee, R. F. Gustducin is a taste- metabotropic glutamate receptors. Neuron 8, 169179 (1992).
cell-specific G protein closely related to the transducins. Nature 357, 563569 34. Bradley, S. R., Levey, A. I., Hersch, S. M. & Conn, P. J. Immunocytochemical
(1992). localization of group III metabotropic glutamate receptors in the
4. McLaughlin, S. K., McKinnon, P. J., Spickofsky, N., Danho, W. & Margolskee, hippocampus with subtype-specific antibodies. J. Neurosci. 16, 20442056
R. F. Molecular cloning of G proteins and phosphodiesterases from rat taste (1996).
cells. Physiol. Behav. 56, 11571164 (1994). 35. Tanabe, Y. et al. Signal transduction, pharmacological properties and
5. Kusakabe, Y., Abe, K., Tanemura, K., Emori, Y. & Arai, S. GUST27 and closely expression patterns of two rat metabotropic glutamate receptors, mGluR3
related G-protein-coupled receptors are localized in taste buds together with and mGluR4. J. Neurosci. 13, 13721378 (1993).
Gi-protein -subunit. Chem. Senses 21, 335340 (1996). 36. Hettinger, T. P., Frank, M. E. & Myers, W. E. Are the tastes of polycose and
6. Kusakabe, Y. et al. Identification of two alpha-subunit species of GTP-binding monosodium glutamate unique? Chem. Senses 21, 341347 (1996).
proteins, Ga15 and Gaq, expressed in rat taste buds. Biochim. Biophys. Acta 37. Rifkin, B. & Bartoshuk, L.M. Taste synergism between monosodium
1403, 265272 (1998). glutamate and disodium 5-guanylate. Physiol. Behav. 24, 11691172 (1980).
7. Huang, L. et al. G13 colocalizes with gustducin in taste receptor cells and 38. Torii, K. & Cagan, R. H. Biochemical studies of taste sensation. IX.
mediates IP3 responses to bitter denatonium. Nat. Neurosci. 2, 10551062 (1999). Enhancement of L-[3H]glutamate binding to bovine taste papillae by
8. Misaka, T. et al. Taste buds have a cyclic nucleotide-activated channel, 5-ribonucleotides. Biochim. Biophys. Acta 627, 313323 (1980).
CNGgust. J. Biol. Chem. 272, 2262322629 (1997). 39. Caicedo, A., Kim, K. & Roper, S. Glutamate-induced cobalt uptake reveals
9. Kretz, O., Barbry, P., Bock, R. & Lindemann, B. Differential expression of non-NMDA receptors in rat taste cells. J. Comp. Neurol. (in press).
RNA and protein of the three pore-forming subunits of the amiloride- 40. Bigiani, A., Delay, R. J., Chaudhari, N., Kinnamon, S. C. & Roper, S. D.
sensitive epithelial sodium channel in taste buds of the rat. J. Histochem. Responses to glutamate in rat taste cells. J. Neurophysiol. 77, 30483059
Cytochem. 47, 5164 (1999). (1997).
10. Lin, W., Finger, T. E., Rossier, B. C. & Kinnamon, S. C. Epithelial Na+ channel 41. Pin, J. P. & Duvoisin, R. The metabotropic glutamate receptors: structure and
subunits in rat taste cells: localization and regulation by aldosterone. J. Comp. functions. Neuropharmacology 34, 126 (1995).
Neurol. 405, 406420 (1999). 42. Pin, J.-P., Waeber, C., Prezeau, L., Bockaert, J. & Heinemann, S. F. Alternative
11. Ogawa, S. et al. Receptor that leaves a sour taste in the mouth. Nature 395, splicing generates metabotropic glutamate receptors inducing different
555556 (1998). patterns of calcium release in Xenopus oocytes. Proc. Natl. Acad. Sci. USA 89,
12. Ming, D., Ruiz-Avila, L. & Margolskee, R. F. Characterization and 1033110335 (1992).
solubilization of bitter-responsive receptors that couple to gustducin. Proc. 43. Yamaguchi, S. & Nakanishi, S. Regional expression and regulation of
Natl. Acad. Sci. USA 95, 89338938 (1998). alternative forms of mRNAs derived from two distinct transcription
13. Matsuoka, I., Mori, T., Aoki, J., Sato, T. & Kurihara, K. Identification of novel initiation sites of the rat mGluR5 gene. J. Neurochem. 71, 6068 (1998).
members of G-protein coupled receptor superfamily expressed in bovine 44. Frohman, M. A., Dush, M. K. & Martin, G. R. Rapid production of full-
taste tissue. Biochem. Biophys. Res. Commun. 194, 504511 (1993). length cDNAs from rare transcripts: amplification using a single gene-
14. Abe, K., Kusakabe, Y., Tanemura, K., Emori, Y. & Arai, S. Primary structure specific oligonucleotide primer. Proc. Natl. Acad. Sci. USA 85, 89989002
and cell-type specific expression of a gustatory G protein-coupled receptor (1988).
related to olfactory receptors. J. Biol. Chem. 268, 1203312039 (1993). 45. Towbin, H., Staehelin, T. & Bordon, J. Electrophoretic transfer of proteins
15. Hoon, M. A. et al. Putative mammalian taste receptors: a class of taste-specific from polyacrylamide gels to nitrocellulose sheets: Procedure and some
GPCRs with distinct topographic selectivity. Cell 96, 541551 (1999). applications. Proc. Natl. Acad. Sci. USA 76, 43504354 (1979).

nature neuroscience volume 3 no 2 february 2000 119


2000 Nature America Inc. http://neurosci.nature.com

articles

Identification and characterization


of the high-affinity choline
transporter
Takashi Okuda1, Tatsuya Haga1, Yoshikatsu Kanai2,3, Hitoshi Endou2, Takeshi Ishihara4 and
Isao Katsura4

1 Department of Neurochemistry, Faculty of Medicine, University of Tokyo and CREST of Japan Science and Technology Corporation,
Bunkyo-ku, Tokyo 113-0033, Japan
2 Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
3 PRESTO of Japan Science and Technology Corporation, Mitaka, Tokyo 181-8611, Japan
2000 Nature America Inc. http://neurosci.nature.com

4 Structural Biology Center, National Institute of Genetics and Department of Genetics, School of Life Science, Graduate University for Advanced Studies,
Mishima 411-8540, Japan
Correspondence should be addressed to T.O. (okuda@m.u-tokyo.ac.jp)

In cholinergic neurons, high-affinity choline uptake in presynaptic terminals is the rate-limiting step
in acetylcholine synthesis. Using information provided by the Caenorhabditis elegans Genome
Project, we cloned a cDNA encoding the high-affinity choline transporter from C. elegans (cho-1). We
subsequently used this clone to isolate the corresponding cDNA from rat (CHT1). CHT1 is not
homologous to neurotransmitter transporters, but is homologous to members of the Na+-dependent
glucose transporter family. Expression of CHT1 mRNA is restricted to cholinergic neurons. The
characteristics of CHT1-mediated choline uptake essentially match those of high-affinity choline
uptake in rat brain synaptosomes.

Cholinergic neurons are vital for cognitive functions of the Although complementary DNAs for transporters for major
brain1,2 and are known to be vulnerable in Alzheimers disease3. neurotransmitters such as GABA, noradrenaline, dopamine,
Because cholinergic neurons lack the capacity to synthesize serotonin, glycine and glutamate have been isolated 15, the
choline de novo, their function depends upon choline uptake4. cDNA encoding the high-affinity choline transporter remains
Brain synaptosome studies demonstrate two carrier-medi- to be isolated despite its physiological importance. Moreover,
ated transport systems for choline uptake59. At high concen- the cDNAs encoding cholinergic neuronal markers, choline
trations, choline is transported primarily by a low-affinity and acetyltransferase16 and the vesicular acetylcholine transporter17
Na+-independent system that is inhibited by hemicholinium-3 have been isolated.
(HC3)10 with a high Ki of approximately 50 M. This system Here we report the functional identification and charac-
is thought to be ubiquitously present in cells and to be required terization of a cDNA encoding the high-affinity choline trans-
for phosphatidylcholine synthesis. At low concentrations, porter (cho-1) in the nematode Caenorhabditis elegans. We also
choline is transported by a high-affinity, Na+-dependent sys- describe the isolation and characterization of the rat homolog
tem that is inhibited by HC3 with a low Ki of 10100 nM. The of cho-1, designated CHT1. Identification of this high-affinity
high-affinity system is supposed to be present specifically in choline-transporter molecule may further understanding of
cholinergic neurons, because a substantial proportion of cholinergic neurons and suggest new therapeutic strategies to
choline is converted to acetylcholine only when taken up counter cholinergic-selective neurodegenerative disorders such
through the high-affinity system5,7,8. as Alzheimers disease3,14,18.
The proposal that the high-affinity choline transport system
is unique to cholinergic neurons is supported by the selective loss RESULTS
of the high-affinity choline uptake following depletion of cholin- To isolate the cDNA encoding the high-affinity choline trans-
ergic terminals in a variety of denervation studies8,9. Choline porter, we examined cDNAs predicted by sequences from the C.
uptake is generally believed to be the rate-limiting step in acetyl- elegans Genome Project19 to be members of the Na+-dependent
choline synthesis49. In addition, the high-affinity choline uptake transporter family. Xenopus laevis oocytes were injected with
is regulated by neuronal activity11,12, suggesting that neuronal cRNA prepared from each candidate full-length clone and exam-
activity also regulates acetylcholine synthesis. In Alzheimers dis- ined for induction of high-affinity choline uptake. We used inhi-
ease, cholinergic neurons selectively degenerate. Consistent with bition by 1 M hemicholinium-3 (HC3) as the criteria for
the above hypothesis, the high-affinity choline transporter is high-affinity choline uptake, because this uptake in mammalian
reduced in Alzheimers disease13. On the other hand, some reports brain synaptosomes is completely inhibited by 1 M HC3
suggest upregulation of the high-affinity choline transporter or (K i = 10100 nM) 7,8,10. By contrast, the low-affinity choline
[3H]HC3-binding sites in Alzheimers disease14. uptake, which is ubiquitously distributed, is inhibited only at

120 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 1. Identification, expression and localization of


a b c C. elegans cho-1. (a) [3H]choline uptake in Xenopus
oocytes injected with C48D1.3 cRNA or water.
Black and gray bars indicate choline uptake values in
the absence and presence of 1 M hemicholinium-3
Choline uptake (cpm)

(HC3), respectively. Each bar represents the mean


s.e. (n = 68 oocytes). (b) Effects of Na+ on choline
uptake. Black bars indicate choline uptake measured
in the standard medium ([Na+] = 100 mM); gray bars
indicate uptake in the absence of Na+. (Na+ was
replaced by Li+.) (c) Inhibition of choline uptake by
HC3. (d) Distribution of neurons that express cho-
1::gfp in the nervous system in C. elegans. An image
of a L1 larvae of N2 carrying an extrachromosomal
array of cho-1::gfp reporter constructs. The arrow-
Water C48D1.3 Water C48D1.3 log[HC3] (M) head indicates the nerve ring. In the ventral nerve
cord, GFP is expressed only in the cholinergic motor
d neurons. However, some DA and DB neurons do
not express GFP, probably because of the loss of the
extrachromosomal array. Scale bar, 50 m.
2000 Nature America Inc. http://neurosci.nature.com

acids with 51% identity and 70% similarity to


cho-1 (Fig. 2a). The rat cDNA clone was desig-
nated CHT1. The amino-acid sequence of
CHT1 has significant homology with members
of the family of Na+-dependent glucose trans-
porters20 (2025%; Fig. 2b), but no significant
homology with those of the choline transporter
of yeast21, the mammalian creatine transporter
originally identified as a high-affinity choline
transporter22 or any known neurotransmitter
higher concentrations of HC3 (Ki of approximately 50 M). One transporters15. The topological model predicted for CHT1 is essen-
cDNA clone corresponding to the predicted gene C48D1.3 tially the same as that of C. elegans CHO-1 (Fig. 2c).
induced a small but significant choline uptake that was inhibited Expression of CHT1 mRNA was examined by Northern blots
by 1 M HC3 (Fig. 1a). This HC3-sensitive uptake was Na + and insitu hybridization. Northern blots of various rat tissues
dependent, and the apparent Ki for HC3 was estimated to be 50 for CHT1 mRNA showed expression of 5-kb transcripts
nM (Fig. 1b and c). Hence we designated this clone cho-1 (high- (Fig. 3a). High mRNA expression was observed in basal fore-
affinity choline transporter-1). brain, brainstem and spinal cord, and low expression was seen
A comparison of the cDNA and the genomic sequence indi- in striatum. Each of these regions contains cholinergic neurons.
cates that the cho-1 gene is composed of nine exons (data not In contrast, no transcripts were detected in other areas of brain or
shown). The cho-1 cDNA sequence predicts a protein of 576 the non-neuronal tissues examined. Consistent with the North-
amino acids (Fig. 2a). A search of available databases with the ern blots, insitu hybridization demonstrated expression of CHT1
amino-acid sequence revealed weak but significant homology mRNA in major cholinergic cell groups examined, including the
with members of the Na+-dependent glucose transporter fami- striatum, ventral spinal cord and scattered cell groups in the basal
ly20 (Fig. 2b). Analysis of hydrophobicity and comparison with forebrain (Fig. 3b and c). This distribution was virtually identi-
other transporters suggests a topological configuration contain- cal to those for choline acetyltransferase23 and vesicular acetyl-
ing twelve transmembrane regions (Fig. 2c). choline transporter24 mRNA, indicating that CHT1 mRNA was
To identify the cells that express cho-1, we introduced the expressed exclusively in cholinergic neurons.
green fluorescent protein (GFP) gene linked to the 5.1-kb Choline uptake in Xenopus oocytes injected with CHT1 cRNA
upstream region of cho-1 gene into worms. Expression of GFP was two- to fourfold greater than the background uptake in con-
was detected in the nervous system, including the nerve ring and trol oocytes injected with water (Fig. 4a). Choline uptake medi-
cholinergic motor neurons of the ventral nerve cord, supporting ated by CHT1 saturated with increasing concentrations of choline
the idea that cho-1 encodes the high-affinity choline transporter with a Km of 2.2 0.2 M (n = 3; Fig. 4b). The apparent Km of
in cholinergic neurons (Fig. 1d). choline for endogenous uptake in control oocytes was greater
We next focused on the vertebrate homolog of cho-1. A data- than 10 M (data not shown). Choline uptake mediated by
base search using the predicted amino-acid sequence of cho-1 iden- CHT1 was completely inhibited by less than 0.1 M HC3 with
tified an entry (GenBank accession number AQ316435) in the a Ki of 25 nM (Fig. 4c). By contrast, choline uptake was only
human genomic survey sequence (GSS). Based on the sequence slightly inhibited by 10 M HC3 in control oocytes. Examina-
homology between cho-1 and this human genomic clone, we tion of the ionic dependency of CHT1-mediated choline uptake
amplified a cDNA fragment from the rat spinal cord cDNA using revealed that the uptake is not only Na+ dependent, but also Cl
degenerate PCR primers. This fragment was used to screen a rat dependent (Fig. 4d). These results show that CHT1-mediated
spinal cDNA library and to isolate positive clones. The sequence choline uptake had the expected characteristics of the high-affin-
of the longest open reading frame predicted a protein of 580 amino ity choline uptake in mammalian synaptosomes studies58 (high

nature neuroscience volume 3 no 2 february 2000 121


2000 Nature America Inc. http://neurosci.nature.com

articles

a b

c
2000 Nature America Inc. http://neurosci.nature.com

Fig. 2. Deduced amino-acid sequence and topological model for the high-affinity choline transporter. (a) Alignment of the predicted amino-
acid sequence of rat CHT1 and C. elegans CHO-1. Numbers in the right column correspond to amino-acid residues. Identical residues are in
bold. The putative transmembrane domains IXII are underlined. (b) Phylogenetic tree of Na+-dependent glucose transporter family. The tree
was constructed by the neighbor-joining method37 using the CLUSTALW program of National Institute of Genetics (Mishima, Japan). The per-
cent amino-acid identity between rat CHT1 and related proteins is shown to the right. Abbreviations: rbSNST1, rabbit sodium/nucleoside
cotransporter 1 (SwissProt accession number P26430); rSGLT1, rat sodium/glucose cotransporter 1 (P53790); hSMIT1, human sodium/myo-
inositol cotransporter 1 (P53794); putP, E. coli sodium/proline symporter (P07117); panF, E. coli sodium/pantothenate symporter (P16256);
rNIS, rat Na/I symporter (PIR accession number S68513); rSMVT, rat Na+-dependent vitamin transporter (PRF accession number 2413365A).
(c) Predicted topology of rat CHT1 in the membrane. Circles represent individual amino acids. Filled circles indicate residues identical (black)
or highly conserved (gray) with C. elegans CHO-1, and open circles indicate divergent residues. Branched lines indicate potential N-linked gly-
cosylation sites. Circled P indicates consensus site for protein kinase C phosphorylation.

affinity for choline, high sensitivity to HC3 and dependence on transporter found in cholinergic nerve endings. We isolated the
Na+ and Cl ions). cDNA clone for the high-affinity choline transporter by using
We also examined [3H]HC3 binding activity of membranes information provided by the C. elegans Genome Project19 and
prepared from COS7 cells, which were transfected with CHT1 systematically expressing putative transporters one by one in
cDNA or vector only (control). Na +-dependent binding of Xenopus oocytes. We then extended this use of C. elegans genom-
[3H]HC3 was detected in membranes from CHT1-transfected ic sequences to isolate a mammalian homolog. A similar strategy
cells but not in membranes from control cells. (Fig. 5a). The equi- may be applicable to expression cloning of unidentified cDNAs of
librium dissociation constant (Kd) was estimated to be 1.6 0.2 biological importance.
nM (n = 3; Fig. 5b), which is similar to the values reported for Our results may explain why the high-affinity choline trans-
brain synaptosomes10,25,26. Specific binding of [3H]HC3 was dis- porter was not previously cloned. First, CHT1 has no significant
placed by tenfold lower concentrations of choline than of acetyl- homology with members of neurotransmitter transporter fami-
choline (Fig. 5c). These results indicate that CHT1 is not only a ly, precluding the use of homology-based cloning to obtain CHT1
high-affinity choline transporter, but also a HC3 binding site. cDNA. CHT1 is not expected to belong to the Na+-dependent glu-
cose transporter family because the Na+-dependent glucose trans-
DISCUSSION porter depends on Na+ but not Cl ions20, whereas the high-affinity
In this study, we identified and characterized cDNAs encoding choline uptake in synaptosomes as well as other neurotransmitter
high-affinity choline transporters in C. elegans and rat, and we transporters depends on both Na+ and Cl ions27. Second, the
confirmed that the CHT1 has the same characteristics as the transport rate by CHT1 is not very high compared with the rates

122 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a b

Fig. 3. Expression studies of rat CHT1. (a) Northern analysis of CHT1


mRNA transcripts in rat tissues. The size of standards (0.249.5 kb) are c
shown on the left. (b, c) Insitu hybridization of CHT1 transcripts in rat
brain and spinal cord. Bright-field micrographs of cryosections
hybridized to digoxigenin-labeled antisense cRNA probe. CHT1 mRNA
2000 Nature America Inc. http://neurosci.nature.com

transcripts were detected in the vertical and horizontal limbs of the


diagonal band (VDB, HDB), medial septal nucleus (MS), caudate and
putamen (CPu) and olfactory tubercle (Tu; b). In the spinal cord, expres-
sion was detected in the ventral horn (VH; c). Adjacent sections
hybridized with a probe for the vesicular acetylcholine transporter
showed essentially the same pattern (data not shown). Sense probe
yielded no signal. Scale bars, 1 mm.

of the high-capacity, low-affinity transporters present in Xenopus cholinergic neurons. The human CHT1 gene, however, occurred
oocytes or cultured cells, possibly explaining previous failures to at a locus different from the one for ChAT and VAChT (data not
detect the transporter by expression cloning. shown). It will be interesting to determine the similarities of the
Choline acetyltransferase (ChAT) and vesicular acetylcholine CHT1 promoter region and CHT1 mRNA distribution to those
transporter (VAChT) are two proteins identified as markers for for ChAT and VAChT.
cholinergic neurons. Their genes are present at the same locus, Earlier studies using mammalian synaptosomes show that
with the VAChT gene positioned within the first intron of the choline taken up through the high-affinity uptake system is effi-
ChAT gene, and their expression is regulated by the same pro- ciently converted to acetylcholine, although choline taken up
moter28,29. We identified CHT1 as a third marker protein for through the low-affinity system is not significantly converted to

a b c
CHT1-mediated choline uptake

(fmol/oocyte/30 min)
(pmol/oocyte/30 min)
(pmol/oocyte/30 min)
[3H]Choline uptake

Choline uptake

Water CHT1 [Choline] (M)


log[HC3] (M)

Fig. 4. Functional expression of rat CHT1 in Xenopus oocytes. d


(a) [3H]choline uptake into Xenopus oocytes injected with CHT1 cRNA or
water. Black and gray bars indicate choline uptake measured in standard
(pmol/oocyte/30 min)

medium containing 100 mM NaCl or LiCl, respectively. Each bar represents the
Choline uptake

mean s.e. (n = 68 oocytes). (b) Effect of choline concentrations on CHT1-


mediated choline uptake. The uptake in water-injected oocytes was subtracted
from that in cRNA-injected oocytes, yielding CHT1-mediated choline uptake.
The uptake was fitted to the Michaelis-Menten curve. (c) Inhibition of choline
uptake by hemicholinium-3 (HC3). (d) Na+ and Cl dependence of choline
uptake. Gray and black bars indicate choline uptake into water- or cRNA-
injected oocytes, respectively. NaCl (100 mM) in the standard uptake solution
was replaced by equimolar amounts of the indicated salts. NaCl LiCl KCl RbCl NaBr NaF Nal

nature neuroscience volume 3 no 2 february 2000 123


2000 Nature America Inc. http://neurosci.nature.com

articles

a b c

Specific Z[3H]HC3 binding

Specific [3H]HC3 binding


(fmol/mg protein)

(fmol/mg protein)
[3H]HC3 binding

(%)
pcDNA pcDNACHT1 Free [3H]HC3 (nM)
log[Competitor] (M)

Fig. 5. Functional expression of rat CHT1 in COS7 cells. (a) Binding of [3H]HC3 to membranes prepared from COS7 cells transfected with
CHT1 cDNA or vector (pcDNA3.1) only. (b) Saturation analysis of specific [3H]HC3 binding. (c) Displacement of specific [3H]HC3 binding by
HC3, choline (Cho) and acetylcholine (ACh). Acetylcholine was tested in the presence of 1 M physostigmine.
2000 Nature America Inc. http://neurosci.nature.com

acetylcholine5,7,8. These findings suggest a close relationship site. NLS and gfp sequences were amplified from pPD104.53 (provided by
between high-affinity choline uptake and acetylcholine synthe- A. Fire). To prepare the 5.1-kb region upstream from the cho-1 transla-
sis30. The cloning of CHT1 provides a means to directly examine tion initiation site, we used a PCR primer that was designed to include the
if and how CHT1 and ChAT interact. in-frame third amino-acid codons of cho-1. The rol-6 (su1006) marker
and the constructed DNA were co-injected into the gonads of animals
Cholinergic neurons are crucial in learning and memory1,2, as described36.
and cholinergic deficits are correlated with the severity of
dementia3. High-affinity choline uptake is the rate-limiting Northern analysis. Poly(A)+RNA (6 g) prepared from different rat tissues
step in acetylcholine synthesis, and its activity is regulated by was resolved by formaldehyde-agarose gel electrophoresis, transferred
neuronal activity11,12 and other stimuli3133. Furthermore, high- onto nylon membranes and hybridized to a random-primed [32P]-labeled
affinity choline uptake or HC3 binding are upregulated in cDNA fragment of CHT1 for 16 h at 42C in 50% formamide, 5 SSPE,
Alzheimers disease14. The cloning of CHT1 may be important 5 Denhardts solution, 0.5% SDS and 100 g per ml salmon sperm DNA.
in discovering the molecular mechanisms underlying these reg- The filters were washed to a final stringency of 0.1 SSPE, 0.1% SDS at
ulations and in developing new therapeutic strategies for treat- 65C and exposed to film with an enhancing screen for 7 days.
ing Alzheimers disease.
Insitu hybridization. Digoxigenin-labeled antisense transcripts were
synthesized in vitro. Transcripts were alkali-hydrolyzed to an average
METHODS length of 200400 base pairs and hybridized in situ on cryostat sections
Cloning of transporter cDNAs. Candidate cDNAs for the choline trans- (1020 m) of fresh-frozen tissues. For hybridization, sections were incu-
porter were isolated by reverse transcription-PCR from mixed-stage poly(A)+ bated at 45C for 20 h with the labeled cRNA probes (1 g per ml) dis-
RNA, using MarathonTM cDNA Amplification Kit (Clontech, Palo Alto, Cal- solved in 1 Denhardts solution containing 50 mM Tris-HCl (pH 8.0),
ifornia), following the manufacturers protocols. PCR oligonucleotide for- 2.5 mM EDTA, 0.3 M NaCl, 50% formamide, 10% dextran sulphate and
ward primers based on DNA sequences obtained from the C. elegans Genome 1 mg per ml E. coli tRNA. Sections were then washed twice in 2
Project19 were designed to construct putative translation-initiation sites of SSC/50% formamide and once in 1 SSC/50% formamide at 45C. The
the predicted genes. These amplified products were cloned into the Nco I hybridized probes were visualized using anti-digoxigenin Fab fragments
(filled-in) and Not I sites of modified pSPUTK (Strategene, La Jolla, Cali- (Boehringer Mannheim, Mannheim, Germany) and NBT/BCIP sub-
fornia), and the sequences of the inserted DNA were determined. Sequence strate. Sections were developed in substrate solution for 2448 h.
data reported in this paper will appear in the DDBJ/EMBL/GenBank
nucleotide sequence databases with the accession numbers AB030946, Binding assays. [3H]hemicholinium-3 (HC3; 128 Ci per mmol) was
AB030947. The rat CHT1 cDNA was isolated from a rat spinal cord cDNA purchased from NEN Life Science Products (Boston, Massachusetts).
library using the GeneTrapper cDNA Positive Selection System (Gibco BRL, COS7 cells were transiently transfected with pcDNA3.1-CHT1 or
Life Technologies, Rockville, Maryland) and a primer based on the sequence pcDNA3.1 using TransFast Reagent (Promega, Madison, Wisconsin)
of the cDNA fragment, following the manufacturers protocols. After char- and following the manufacturers protocols. To prepare membranes,
acterizing the resulting cDNA clones, we subcloned a positive clone into cells were homogenized in 0.32 M sucrose and centrifuged for 1 h at
pSPUTK and pcDNA3.1+ (Invitrogen, Carlsbad, California). 200,000 g, and the pellet was resuspended. Binding assays were done
essentially as described 26. Specific binding was calculated by sub-
Expression in Xenopus oocytes. SP6 or T7 RNA polymerase in the tracting non-specific binding (determined in the presence of 10 M
presence of cap analog was used to prepare cRNA in vitro. Stage VVI HC3) from total binding. Binding-saturation data were analyzed by
Xenopus laevis oocytes were injected with 2030 ng capped cRNA. The nonlinear transformation of the calculated specific [3H]HC3 bind-
uptake was assayed essentially as described34. Choline uptake (23 d ing to generate a Kd value.
after injection) was measured by incubating 68 oocytes for 3045
min with [3H]choline chloride in 750 l standard medium (100 mM ACKNOWLEDGEMENTS
NaCl, 2 mM KCl, 1 mM MgCl2, 1 mM CaCl2, 10 mM HEPES, 5 mM We thank Y. Iino for the C. elegans N2 strain, Y. Kobayashi for help with basal
Tris, pH 7.4). Oocytes were solubilized with 10% SDS, and the [3H] forebrain preparations, A. Fire for pPD104.53, S. Yamashita for yeast choline-
content was measured by liquid scintillation counting. transporter cDNA, T. Suzuki and Y. Kirino for Torpedo electric lobe and its cDNA
GFP expression construct. The cho-1::gfp transcriptional fusion was con- library, Y. Koyama for laterodorsal tegmental nucleus, K. Kameyama for
structed by PCR essentially as described35. The green fluorescent protein suggestions and D. Saffen for English corrections. This work was supported by
gene preceded by a nuclear localization signal (NLS) sequence was insert- grants from Japan Science and Technology Corporation (CREST) and the Ministry
ed in-frame, 3 amino acids downstream of the cho-1 translation start of Japan Society for the Promotion of Science (Research for Future Program).

124 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

RECEIVED 1 NOVEMBER; ACCEPTED 23 NOVEMBER 1999 21. Nikawa, J., Hosaka, K., Tsukagoshi, Y & Yamashita, S. Primary structure of
the yeast choline transport gene and regulation of its expression. J. Biol.
Chem. 265, 1599616003 (1990).
1. Winkler, J. et al. Essential role of neocortical acetylcholine in spatial memory. 22. Schloss, P., Mayser, W. & Betz, H. The putative rat choline transporter
Nature 375, 484487 (1995). CHOT1 transports creatine and is highly expressed in neural and muscle-
2. Dutar, P., Bassant, M. H., Senut, M. C. & Lamour, Y. The septohippocampal rich tissues. Biochem. Biophys. Res. Commun. 198, 637645 (1994).
pathway: structure and function of a central cholinergic system. Physiol. Rev. 23. Oh, J. D. et al. Cholinergic neurons in the rat central nervous system
75, 393427 (1995). demonstrated by in situ hybridization of choline acetyltransferase mRNA.
3. Bierer, L. M. et al. Neurochemical correlates of dementia severity in Neuroscience 47, 807822 (1992).
Alzheimers disease: relative importance of the cholinergic deficits. J. 24. Roghani, A. et al. Molecular cloning of a putative vesicular transporter for
Neurochem. 64, 749760 (1995). acetylcholine. Proc. Natl. Acad. Sci. USA 91, 1062010624 (1994).
4. Tucek, S. Regulation of acetylcholine synthesis in the brain. J. Neurochem. 44, 25. Vickroy, T. W., Roeske, W. R. & Yamamura, H. I. Sodium-dependent high-
1124 (1985). affinity binding of [3H]hemicholinium-3 in the rat brain: a potentially
5. Haga, T. Synthesis and release of [14C]acetylcholine in synaptosomes. J. selective marker for presynaptic cholinergic sites. Life Sci. 35, 23352343
Neurochem. 18, 781798 (1971). (1984).
6. Yamamura, H. I. & Snyder, S. H. Choline: high-affinity uptake by rat brain 26. Sandberg, K. & Coyle, J. T. Characterization of [3H]hemicholinium-3
synaptosomes. Science 178, 626628 (1972). binding associated with neuronal choline uptake sites in rat brain
7. Haga, T. & Noda, H. Choline uptake systems of rat brain synaptosomes. membranes. Brain Res. 348, 321330 (1985).
Biochim. Biophys. Acta 291, 564575 (1973). 27. Kuhar, M. J. & Zarbin, M. A. Synaptosomal transport: a chloride dependence
8. Kuhar, M. J. & Murrin, L. C. Sodium-dependent, high-affinity choline for choline, GABA, glycine and several other compounds. J. Neurochem. 31,
uptake. J. Neurochem. 30, 1521 (1978). 251256 (1978).
9. Kuhar, M. J., Sethy, V. H., Roth, R. H. & Aghajanian, G. K. Choline: selective 28. Erickson, J. D. et al. Functional identification of a vesicular acetylcholine
accumulation by central cholinergic neurons. J. Neurochem. 20, 581593 transporter and its expression from a cholinergic gene locus. J. Biol. Chem.
(1973). 269, 2192921932 (1994).
2000 Nature America Inc. http://neurosci.nature.com

10. Happe, H. K. & Murrin, L. C. High-affinity choline transport sites: use of 29. Bejanin, S., Cervini, R., Mallet, J. & Berrard, S. A unique gene organization
[3H]hemicholinium-3 as a quantitative marker. J. Neurochem. 60, 11911201 for two cholinergic markers, choline acetyltransferase and a putative
(1993). vesicular transporter of acetylcholine. J. Biol. Chem. 269, 2194421947
11. Simon, J. R. & Kuhar, M. J. Impulse-flow regulation of high affinity choline (1994).
uptake in brain cholinergic nerve terminals. Nature 255, 162163 (1975). 30. Barker, L. A. & Mittag, T. W. Comparative studies of substrates and
12. Murrin, L. C. & Kuhar, M. J. Activation of high-affinity choline uptake in inhibitors of choline transport and choline acetyltransferase. J. Pharmacol.
vitro by depolarizing agents. Mol. Pharmacol. 12, 10821090 (1976). Exp. Ther. 192, 8694 (1975).
13. Pascual, J. et al. High-affinity choline uptake carrier in Alzheimers disease: 31. Vogelsberg, V., Neff, N. H. & Hadjiconstantinou, M. Cyclic AMP-mediated
implications for the cholinergic hypothesis of dementia. Brain Res. 552, enhancement of high-affinity choline transport and acetylcholine synthesis
170174 (1991). in brain. J. Neurochem. 68, 10621070 (1997).
14. Bissette, G., Seidler, F. J., Nemeroff, C. B. & Slotkin, T. A. High affinity choline 32. Beeri, R. et al. Enhanced hemicholinium binding and attenuated dendrite
transporter status in Alzheimers disease tissue from rapid autopsy. Ann. NY branching in cognitively impaired acetylcholinesterase-transgenic mice. J.
Acad. Sci. 777, 197204 (1996). Neurochem. 69, 24412451 (1997).
15. Nelson, N. The family of Na+/Cl neurotransmitter transporters. J. 33. Kar, S. et al. Amyloid -peptide inhibits high-affinity choline uptake and
Neurochem. 71, 17851803 (1998). acetylcholine release in rat hippocampal slices. J. Neurochem. 70, 21792187
16. Berrard, S. et al. cDNA cloning and complete sequence of porcine choline (1998).
acetyltransferase: in vitro translation of the corresponding RNA yields an 34. Kanai, Y. & Hediger, M. A. Primary structure and functional
active protein. Proc. Natl. Acad. Sci. USA 84, 92809284 (1987). characterization of a high-affinity glutamate transporter. Nature 360,
17. Alfonso, A. et al. The Caenorhabditis elegans unc-17 gene: a putative vesicular 467471 (1992).
acetylcholine transporter. Science 261, 617619 (1993). 35. Cassata, G. et al. Rapid expression screening of Caenorhabditis elegans
18. Wurtman, R. J. Choline metabolism as a basis for the selective vulnerability of homeobox open reading frames using a two-step polymerase chain reaction
cholinergic neurons. Trends Neurosci. 15, 117122 (1992). promoter-gfp reporter construction technique. Gene 212, 127135 (1998).
19. The C. elegans Sequencing Consortium. Genome sequence of the nematode 36. Mello, C. C., Kramer, J. M., Stinchcomb, D. & Ambros, V. Efficient gene
C. elegans: a platform for investigating biology. Science 282, 20122018 transfer in C. elegans extrachromosomal maintenance and integration of
(1998). transforming sequences. EMBO J. 10, 39593970 (1991).
20. Hediger, M. A. & Rhoads, D. B. Molecular physiology of sodium-glucose 37. Saitou, N. & Nei, M. The neighbor-joining method: a new method for
cotransporters. Physiol. Rev. 74, 9931026 (1994). reconstructing phylogenetic trees. Mol. Biol. Evol. 4, 406425 (1987).

nature neuroscience volume 3 no 2 february 2000 125


2000 Nature America Inc. http://neurosci.nature.com

articles

Receptors with opposing functions


are in postsynaptic microdomains
under one presynaptic terminal
Guoshan Tsen1, Brian Williams1, Pauline Allaire2, Yu-Dong Zhou1, Ognian Ikonomov3,
Ivanela Kondova4 and Michele H. Jacob1

1 Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave., Boston, Massachusetts 02111, USA
2 Blackstone-Millville Regional High School, 175 Lincoln St., Blackstone, Massachusetts 01504, USA
3 Dept. of Psychiatry, Wayne State University Med. Ctr., 540 E. Canfield St., Detroit, Michigan 48201, USA
4 Department of Infectious Disease, Tufts University Veterinary School, 200 Westboro Rd., Grafton, Massachusetts 01536, USA
2000 Nature America Inc. http://neurosci.nature.com

The first two authors contributed equally to this work.


Correspondence should be addressed to M.H.J. (mjacob@opal.tufts.edu)

Fast excitatory synaptic transmission through vertebrate autonomic ganglia is mediated by postsynaptic
nicotinic acetylcholine receptors (nAChRs). We demonstrate a unique postsynaptic receptor microhetero-
geneity on chick parasympathetic ciliary ganglion neuronsunder one presynaptic terminal, nAChRs and
glycine receptors formed separate but proximal clusters. Terminals were loaded with [3H]glycine via the
glycine transporter-1 (GlyT-1), which localized to the cholinergic presynaptic terminal membrane;
depolarization evoked [3H]glycine release that was calcium independent and blocked by the GlyT-1
inhibitor sarcosine. Ganglionic synaptic transmission mediated by nAChRs was attenuated by glycine.
Coexistence of separate clusters of receptors with opposing functions under one terminal contradicts
Dales principle and provides a new mechanism for modulating synaptic activity in vivo.

Nicotinic acetylcholine receptors mediate fast excitatory synap- RESULTS


tic transmission through all ganglia in the vertebrate periph- On chick ciliary neurons, nAChRs are localized predominantly
eral nervous system (PNS). It is thought that this synapse in the postsynaptic membrane (Fig. 1)1113. We showed this at
functions as a simple relay, and that modulation occurs cen- the ultrastructural level with immunocytochemistry using mon-
trally through the integration of excitatory and inhibitory oclonal antibody (mAb) 35 to nAChRs, visualized with horse-
inputs to the preganglionic neurons. However, functional radish peroxidase (HRP). In this study, nAChR refers strictly to
inhibitory glycine receptors (GlyRs) were found on embry- the 3-containing nAChRs, which are concentrated in the post-
onic chick parasympathetic ciliary ganglion (CG) neurons1, synaptic membrane and are specifically recognized by mAb 35,
raising questions as to their spatial distribution and physio- and does not include the 7-containing nAChRs, which are
logical significance. These issues are particularly intriguing, perisynaptic, excluded from the postsynaptic membrane and not
as each ciliary neuron is innervated by only one presynaptic recognized by mAb 35 (refs. 13-16). We defined synapses by char-
terminal, a large calyx-type ending2,3. Moreover, the terminal acteristic morphological featuresa thickened and parallel
is cholinergic and is derived from the sole source of presy- arrangement of the pre-and postsynaptic membranes, an accu-
naptic input to the CG, the accessory oculomotor nucleus mulation of synaptic vesicles adjacent to the presynaptic density,
(AON)28. Multiple synaptic contacts are formed in the region an enhanced postsynaptic density and a widened synaptic cleft.
of apposition between the calyx ending and the postsynaptic Concentrations of nAChRs are found in patches of postsynaptic
neuron. According to the current interpretation of Dales prin- membrane demarcated by the postsynaptic density along their
ciple, all of the active zones of the presynaptic terminals cytoplasmic face and lying opposite the presynaptic density of
derived from one axon release the same combination of neu- the active zone. However, labeling for nAChRs was not found in
rotransmitters, and all of the underlying specialized postsy- all of the specialized postsynaptic membrane microregions under
naptic membrane regions on one neuron are predicted to one calyx ending (Fig. 1a). This selective staining pattern was
contain the same receptor types9,10. We show here that this is unlikely to result from limited penetration of labeling reagents, as
not the case. We demonstrate a unique postsynaptic receptor unlabeled postsynaptic membrane microregions were close to
microheterogeneity: under a single presynaptic terminal, exci- labeled ones.
tatory nAChR clusters and separate but proximal inhibitory GlyRs were concentrated in similar specialized postsynaptic
GlyR clusters are localized in discrete membrane membrane microdomains on late-stage embryonic ciliary neu-
microdomains. These findings are surprising, considering that rons as shown by immunolabeling with either mAb 4a or mAb
these receptors respond to different fast-acting transmitters 2b and ultrastructural analysis (Fig. 1b). On each neuron exam-
and have opposing actions. ined, some, but not all, membrane microregions demarcated by

126 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 1. Ultrastructural localization of nAChRs, GlyRs


and gephyrin at discrete postsynaptic membrane a
microdomains under the calyx-type presynaptic ter-
minal on E1517 chick ciliary neurons in vivo. The
three components were present at only a subset of
the postsynaptic membrane microdomains demar-
cated by the postsynaptic density along their cyto-
plasmic faces and lying opposite the synaptic vesicle
accumulations at the active zone (labeled synapse, b c
black arrowhead; unlabeled synapse, white arrow-
head). (a) nAChRs detected by immunolabeling with
mAb 35. (b) GlyRs labeled with mAb 2b. (c) gephyrin
labeled with mAb 7a (visualized with HRP). Scale bar,
1 m. (d) Schematic representation of the relative
distribution of nAChRs, GlyRs and gephyrin at spe-
cialized postsynaptic membrane microdomains under d
the calyx terminal (nt; synaptic vesicles, sv; short den-
drites, d; based on data in Figs. 1 and 2).
2000 Nature America Inc. http://neurosci.nature.com

the postsynaptic density were labeled for GlyRs.


The presence of both GlyR and nAChR accu-
mulations in postsynaptic membrane
microdomains under the cholinergic calyx ter-
minal was unexpected.
As gephyrin is a peripheral membrane pro-
tein known to codistribute with GlyRs at synapses and is required clusters were present in approximately equal proportions on all
for GlyR clustering on rodent CNS neurons1820, we used it as a CG neurons at stages after pre-and postganglionic synapse for-
positive control to examine the relative distribution of GlyRs and mation, ranging from late-stage embryonic ages to reproduc-
nAChRs on avian ciliary neurons. We first determined that tively mature adults. Functional GlyRs were present on CG
gephyrin was expressed in avian CG neurons and that the avian neurons at earlier embryonic ages as well, but their expression
and rodent sequences were highly conserved, suggesting that was developmentally delayed relative to functional nAChRs1. As
mAbs to rodent gephyrin could be used for localization studies independent confirmation of the GlyR and gephyrin colocaliza-
in chick. Specifically, we isolated the full-length gephyrin coding tion results, we showed by the yeast two-hybrid genetic assay that
sequence from a chick brain cDNA library (GenBank accession chick gephyrin specifically interacts with the chick GlyR -sub-
number AF174130). The avian and rodent sequences showed unit, but not with nAChR subunits (Fig. 2c). We tested for
87% identity at the nucleotide level and 98% identity at the gephyrin interactions with the major cytoplasmic loop of the
amino-acid level. We then examined gephyrin mRNA expression receptor subunits, as the gephyrin binding motif in the rodent
in the chick CG by using RT-PCR amplification of total gan- GlyR is a linear 18-amino-acid (aa) sequence in the -subunit
glionic RNA. Because five alternatively spliced variants of long cytoplasmic loop23,24. We established that this 18-aa sequence
gephyrin are expressed in the rodent in a tissue-specific pattern21, was completely conserved in the chick GlyR -subunit (GenBank
we used seven different pairs of gephyrin-specific primers to flank accession number AF181717) as determined by RT-PCR ampli-
all of the known alternatively spliced cassettes identified in rodent fication of CG total RNA using specific primers corresponding
cDNA (see Methods). We exclusively detected the gephyrin to the mammalian GlyR -subunit sequence. The gephyrin bind-
mRNA isoform containing the C2 cassette in the chick CG, as ing motif was absent from neuronal nAChR subunits. Only yeast
determined by size and sequence analysis of the PCR amplifica- transformants co-expressing the chick GlyR -subunit loop and
tion products (n = 14 separate experiments; data not shown). gephyrin showed transcriptional activation of the two reporter
This is also the dominant isoform expressed in the rodent CNS21. genes, HIS3 and LacZ (Fig. 2c). Altogether, the colocalization
Immunocytochemical labeling of avian ciliary neurons with studies revealed a synaptic microheterogeneity consisting of sep-
anti-gephyrin mAb 5a or mAb 7a20,22 and ultrastructural analysis arate but proximal clusters of nAChRs and of GlyRs in discrete
demonstrated gephyrin distribution similar to that of the recep- postsynaptic membrane microdomains under one presynaptic
tors, with labeling present at only a subset of the specialized post- terminal (Fig. 1d). The protein-interaction data suggest that dif-
synaptic membrane microdomains under the calyx (Fig. 1c). A ferent molecules organize the nAChR and GlyR clusters in the
minor difference was that gephyrin staining was concentrated at distinct postsynaptic microdomains.
the postsynaptic density rather than at the postsynaptic mem- To determine whether this synaptic heterogeneity was a
brane. To determine the synaptic distribution of nAChRs, GlyRs unique feature of a calyx-type synapse, we examined the choroid
and gephyrin relative to one another, we used immunofluores- neuron subpopulation in the chick CG, as they are innervated
cent double labeling with laser-scanning confocal microscopy. In by multiple typical bouton-type endings. Each choroid neuron
both late-stage embryonic and adult CGs, GlyR (red) and receives inputs from two to three preganglionic neurons in the
gephyrin (green) clusters were colocalized on the neuron surface, AON3. Separate clusters of nAChRs and of GlyRs (and the GlyR-
as indicated by the predominance of yellow fluorescent patches associated protein gephyrin) were detected in distinct postsy-
(Fig. 2a). In contrast, nAChR (red) and gephyrin (green) clus- naptic membrane regions under separate bouton terminals
ters did not overlap, as indicated by distinct patches of red or (Fig. 2d). Although there may be separate glycinergic and cholin-
green fluorescence (Fig. 2b). Separate nAChR and GlyR/gephyrin ergic inputs to the choroid neurons, this seems unlikely, as all

nature neuroscience volume 3 no 2 february 2000 127


2000 Nature America Inc. http://neurosci.nature.com

articles

presynaptic terminals to the CG are derived from the AON and To study the functional significance of inhibitory GlyR accu-
are cholinergic28. Thus, as for the calyceal synapse, heteroge- mulations at these largely cholinergic synapses, we investigated
neous microdomains occur under axonal branches terminating in potential sources of endogenous glycine in the CG and conditions
multiple bouton-type endings. that lead to its release. Glycine is present in blood at a high con-
We examined the functional effects of GlyR activation in centration (0.5 mM)29. Glycine transporters (GlyTs) provide an
CG neurons. Using acutely isolated, intact E1517 ganglia, we avenue for glycine uptake, but operate in reverse to release glycine
stimulated the preganglionic nerve and extracellularly record- under certain physiological conditions (such as strong depolar-
ed postsynaptic responses with or without exogenous glycine ization and high internal sodium concentration)30. We established
(0.51 mM, Fig. 3). Glycine reduced initial slope of the nAChR- that the two GlyTs, GlyT-1 and GlyT-2, were present in the chick
mediated excitatory postsynaptic potential (EPSP; Fig. 3b and c) CG at both late embryonic and adult stages based on immuno-
by 30% and peak-to-peak amplitude of the postganglionic labeling and uptake studies. The two transporters had different
compound action potential (a.p.) by 65% (data not shown), distributions as determined by anti-GlyT-1 and anti-GlyT-2 spe-
but did not change the preganglionic a.p. volley as indicated cific antisera and immunofluorescence microscopy (Fig. 4a and b).
by the electrical coupling potential (Fig. 3b). This inhibition GlyT-1 (green) was localized predominantly in patches on the
was completely reversed following washout of glycine. Strych- presynaptic terminal surface membrane and colocalized with
nine (110 M), a GlyR antagonist, was not used to test for synaptic-vesicle immunolabeling (red) in the late-stage embry-
GlyR involvement because it also blocks nAChRs on CG neu- onic and adult ganglia (Fig. 4a). GlyT-1 labeling was also present
rons (data not shown)25. Compared with glycine, hexametho- on neuronal cell bodies, but not on Schwann cells. In contrast,
2000 Nature America Inc. http://neurosci.nature.com

nium (0.5 mM), anAChR antagonist, decreased initial slope of GlyT-2 labeling (red) was detected only on Schwann cells (Fig. 4b).
the EPSP by 70% (Fig. 3a and c) and the compound a.p. ampli- It should be noted that the localization of GlyT-1 on presynaptic
tude by 80%, consistent with earlier reports of partial block by nerve terminals and GlyT-2 on Schwann cells in this avian periph-
hexamethonium but complete inhibition of nAChR-mediated eral ganglion is reversed from their typical distribution in the
excitatory responses in the CG by other nicotinic cholinergic mammalian CNS. However, reported exceptions include the reti-
antagonists26. Tetrodotoxin (1 M) abolished all postsynaptic na, where GlyT-1 is expressed on nerve terminals but not on glia,
responses (Fig. 3a and c). Our results were similar to those of and the cerebellum, where GlyT-2 is on both glia and terminal
previous electrophysiological studies of CG neurons showing boutons31. In addition, we determined that, in the chick CG, GlyT-
that activation of the chloride-selective GlyR channel drives 1 and GlyT-2 facilitated uptake of exogenous glycine. Acutely iso-
the cell toward the chloride equilibrium potential (near the lated, intact E1517 ganglia were treated with collagenase to
resting potential and away from a.p. threshold) and that acti- increase reagent access, incubated with [3H]glycine (1 M), rinsed
vation of the GABAA receptor, another chloride-selective chan- and processed either for scintillation counting or light-micro-
nel, completely blocks synaptic transmission through the CG at scopic autoradiographic analysis. In addition, the intact ganglia
the embryonic age tested here1,27,28. In sum, GlyR activation were incubated with FM1-43 to label synaptic vesicles and there-
inhibits excitatory synaptic activity in the late-stage embryon- by identify presynaptic terminals. Adjacent sections were processed
ic chick CG. for fluorescence or autoradiography. We observed dense clusters

a b c d

Yeast two-hybrid assay


Gephyrin interaction with

Fig. 2. Segregation of nAChR clusters from clusters of GlyRs and their


directly associated protein gephyrin in postsynaptic membrane
microdomains, including those under separate cholinergic bouton-type
terminals on CG choroid neurons. (a) Confocal micrograph of a single
E21 CG neuron showing colocalization of GlyR (red) and gephyrin (green) clusters on the neuron surface (predominance of yellow fluorescent
patches) by immunofluorescent double labeling. In addition, there were a few gephyrin clusters (green patches) lacking in GlyR staining. Either these
sites may represent a distinct subset of gephyrin clusters or the plane of section in these regions may favor the intracellular postsynaptic density
(gephyrin localization) and show less of the postsynaptic surface membrane (GlyR localization). Only 2 optical sections (each 0.5 m) were summed
in this image; including additional optical sections resulted in complete overlap, yielding only yellow patches. (b) In contrast, nAChR (red) and gephyrin
(green) clusters did not overlap on the E21 CG neuron surface (distinct red or green patches). Scale bar, 10 m. (c) Yeast two-hybrid genetic assay
showing chick gephyrin specifically interacts with chick GlyR -subunit long cytoplasmic loop, but not with nAChR subunit loops. For the -gal assay,
+++, blue color within 1 h; , not blue after overnight. For the HIS3 assay, +, overnight growth; no growth on histidine-deficient media. (d) Electron
micrograph showing nAChRs at the specialized postsynaptic membrane microdomains under a subset of bouton-type endings (nt) on a choroid neu-
ron in the E1517 CG in vivo (black arrowhead, labeled synapse; white arrowhead, unlabeled synapse). Scale bar, 1.2 m.

128 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a b

0.2 mV

3 ms
0.1 mV

Fig. 3. Inhibitory GlyR response in CG neurons. (a) Schematic showing 1 ms


the position of the recording electrode used to measure postsynaptic
responses extracellularly following direct electrical stimulation of the
preganglionic nerve in an acutely isolated, intact E1517 CG. Top trace
2000 Nature America Inc. http://neurosci.nature.com

(control) shows a typical postsynaptic response composed of the cou-


pling potential (); due to direct electrical connection between pre- and c
postsynaptic cells, followed with a slight delay by the chemically medi-
ated field EPSP ()4,5. Bath-applied hexamethonium (0.5 mM) decreased
the initial slope of the field EPSP, with no detectable alteration in the
coupling potential. Tetrodotoxin (1 M) abolished all postsynaptic
responses. (b) Representative trace showing that exogenous glycine (0.5
mM) reduced the initial slope of the nAChR-mediated EPSP (), with no
change in the electrical coupling potential (). (c) Histogram of EPSP
initial slope values expressed as the ratio of test over control (mean
s.e.; number of postsynaptic responses measured is shown in parenthe-
ses above the bars; **p < 0.005, ***p < 0.001, Students t-test.

of silver grains representing [3H]glycine uptake over presynaptic attenuated synaptic transmission mediated by nAChRs in the
terminals and Schwann cells, with fewer grains over the neuronal CG. We propose that two distinct release mechanisms, vesicular
cell bodies (Fig. 4c). Pre- and co-incubation of CGs with sarco- acetylcholine release and non-vesicular, transport-mediated
sine, a selective GlyT-1 inhibitor 32,33, dramatically reduced glycine efflux, both occur at the calyceal synapse on ciliary neu-
[3H]glycine uptake in a concentration-dependent manner; at 0.5, rons in vivo and jointly regulate ganglionic transmission.
1 and 5 mM, sarcosine decreased [3H]glycine levels per CG by Our data suggest that GlyT-1 on the cholinergic presynaptic ter-
50%, 60% and 70%, respectively. Moreover, in the presence of 5 minal mediates depolarization-induced glycinergic synaptic trans-
mM sarcosine, fewer silver grains were observed over presynap- mission in the CG. Amino-acid transporters on presynaptic
tic terminals and neuronal somata, whereas label intensity over terminals release high levels of transmitter in response to depolar-
Schwann cells was not detectably altered (Fig. 4d). In compari- ization and can function as a relatively fast release mechanism34. The
son, the addition of 1,000-fold excess cold glycine reduced depolarization protocol required to activate GlyT-1-mediated efflux
[3H]glycine levels per CG by 80% and drastically lowered the in the CG in vivo is not yet defined. It should be noted that nAChRs
number of silver grains over terminals, neuronal somata and composed of 7 subunits are present on the presynaptic calyx end-
Schwann cells (Fig. 4e). Depolarization of these radiolabeled gan- ing and may contribute to depolarization of the terminal35. CG GlyR-
glia with 30 mM KCl induced the release of [3H]glycine (Fig. 4f). rich synaptic microregions do not lack presynaptic vesicles, as
The release was Ca2+ independent and blocked by the GlyT-1 previously reported for synapses with strictly transporter-mediated
antagonist sarcosine or an excess of cold glycine (Fig. 4g), consis- release. These microregions lack classical structural features of
tent with a non-vesicular, transport-mediated mechanism. Alto- inhibitory synapses such as pleomorphic or flattened vesicles; instead,
gether, these results suggest that GlyT-1 on the presynaptic they have small, clear, round presynaptic vesicles, resembling the
terminals probably mediates depolarization-induced glycinergic neighboring cholinergic synaptic microregions30,36. Strikingly simi-
synaptic transmission in the chick CG. lar to the co-existence of vesicular acetylcholine release and trans-
port-mediated glycine efflux suggested by our observations at
DISCUSSION synapses in the CG is the demonstration that retinal starburst
We demonstrated separate clusters of excitatory nAChRs and amacrine cells corelease ACh and GABA via vesicular and carrier-
inhibitory GlyRs in distinct postsynaptic membrane mediated mechanisms, respectively37. The two release mechanisms
microdomains under one presynaptic terminal in vivo. As these differ in their extracellular Ca2+ requirements and in the ranges of
receptors respond to different fast-acting transmitters and gen- membrane voltages over which they are active30,34,37, suggesting that
erally have opposing functions, the presence of both GlyRs and dynamic shifts in the release of transmitters with opposing func-
nAChRs under one presynaptic ending is surprising. Glycine tions from single terminals in the CG are possible.

nature neuroscience volume 3 no 2 february 2000 129


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 4. Glycine transporter localization


and glycine uptake and release in the CG. a b c d
(a) Immunofluorescence micrograph of a
single double-labeled E16 CG neuron
showing GlyT-1 (green) predominantly
on presynaptic terminals, where it par-
tially overlapped with synaptic vesicles
(SV2 antigen, red; overlap, yellow). GlyT-1
was on the neuronal soma, but not on
Schwann cells. (b) In contrast, GlyT-2 e
(red) was localized only on Schwann cells
and did not overlap with synaptic-vesicle
staining (green). Scale bar, 10 m.
(ce) Autoradiograms showing exoge-
f g
nous [3H]glycine uptake sites in the
acutely isolated E1517 CG. (c) Dense
clusters of silver grains were found over

Release per ganglion


Release per ganglion

(ratio test: control)


(ratio test: control)

presynaptic terminals and Schwann cells,


with fewer grains over neuronal soma.
(d) Pre- and co-incubation with sarco-
2000 Nature America Inc. http://neurosci.nature.com

sine, a selective GlyT-1 inhibitor, sharply


reduced the number of silver grains over
terminals and neuronal somata, whereas
the grain density over Schwann cells was
unaltered. (e) Addition of 1,000-fold
excess cold glycine drastically lowered
silver grain levels over all CG areas.
Sections were Nissl stained to mark neu-
rons and Schwann cells. Scale bar, 30 m.
(f) Depolarization (using 30 mM KCl)
induced [3H]glycine release from a pre-
loaded ganglion. (g) Amount of release
was unaltered by adding Cd2+ (0.1 mM) or reducing Ca2+ levels (from 5.4 mM to 0.2 mM), whereas sarcosine (5 mM) and excess cold glycine (1 mM)
decreased high-K+-induced [3H]glycine release. Data are expressed as the mean s.e. of the ratio of the proportion of total ganglionic [3H]glycine
released into the medium in test over control conditions. For each condition, n = 6. *p < 0.02, Students two-tailed t-test.

The receptor arrangement reported here represents an unex- The ultrastructural distribution of gephyrin in the chick CG was estab-
pected degree of complexity and heterogeneity of functionally lished using the anti-rat gephyrin mAbs 5a and 7a (gift from Heinrich
specialized subdomains in the neuronal postsynaptic membrane. Betz, Max-Planck Institute)20,22. To gain intracellular access for gephyrin
Postsynaptic receptor microheterogeneity may have relevance to immunolabeling, freshly dissected E1517 CGs were lightly fixed, deter-
the mammalian CNS as well, as suggested by the demonstration gent-permeabilized with saponin12, incubated with mAb 5a or 7a and
processed as described above.
that two fast-acting excitatory and inhibitory neurotransmitters
are coreleased by a single presynaptic CNS neuron, possibly at Laser-scanning confocal microscopy. Double-labeling immunofluores-
the same synapse, in vitro38. The presence under one terminal of cence and laser-scanning confocal microscopy (Leica TCS) were done as
two distinct receptor types responding to different fast trans- described40,41. Both late-stage embryonic (E15-E21) and adult CGs were
mitters with opposing functions may represent an important analyzed. (Freshly decapitated heads of reproductively mature adult
mechanism for modulating activity at a monosynaptic connec- White Leghorn chickens were obtained from a local fresh poultry sup-
tion in vivo. plier.) The nAChRs were detected by mAb35 and Cy-3-conjugated anti-
rat IgG, gephyrin by mAb 7a and FITC-conjugated anti-mouse IgG, and
GlyRs by mAb 2b and Cy3-conjugated anti-mouse IgG (primary mAbs
METHODS used at 1:1001:200 dilution; secondary IgGs used at 1:1000; Vector Labs;
Electron microscopy. The localization of nAChRs and GlyRs in postsy- Jackson ImmunoResearch Labs, West Grove, Pennsylvania). The anti-
naptic membrane microdomains on the CG neuron surface was estab- rat and anti-mouse IgGs were affinity purified to eliminate cross-reac-
lished at the ultrastructural level. Briefly, CGs were dissected from White tivity with mouse and rat primary mAbs, respectively. Double-labeling
Leghorn chick embryos in ovo (Spafas) at E1517, incubated in 0.1 M with the GlyR and gephyrin mouse mAbs was carried out using a previ-
primary mAb followed by 0.1 M biotinylated secondary antibody and ously described protocol for multiple staining with antibodies raised in
strepavidinbiotinHRP complex (Vector Labs, Burlingame, California), the same species42. As controls to test for specific binding, we omitted
fixed, reacted for peroxidase activity, and processed for electron the first or second primary mAb and reversed the antibody sequence in
microscopy1113. The nAChRs were detected by using rat mAb 35, and separate tests.
GlyRs were detected using mouse mAb 4a or 2b (provided by Heinrich
Betz, Max-Planck Institute for Brain Research, Frankfurt, Germany)17. Light microscopy. To establish the cellular localization of GlyTs in
As a control, CGs were incubated in the absence of primary mAb. Thin E15E17 and adult CGs, frozen ganglionic sections (68 m thickness)12
sections were stained with 5% aqueous uranyl acetate and examined with were labeled with goat anti-GlyT-1 or guinea pig anti-GlyT-2 specific
a Philips CM10 electron microscope (FEI Company, Hillsboro, Oregon). antisera (1:200 and 1:500 dilution, respectively; Chemicon Internation-
Anti-nAChR mAb 35 from a hybridoma isolated by Jon Lindstrom and al, Temecula, California) and FITC-conjugated anti-goat IgG (1:500; Vec-
colleagues (University of Pennsylvania, Philadelphia, Pennsylvania) was tor Labs) and Cy3-conjugated anti-guinea pig IgG (1:1000; Jackson
purified as described39. ImmunoResearch Labs). To determine the distribution of GlyTs relative

130 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

to synapses, synaptic vesicles were stained using anti-SV2 mAb (1:200 Mannheim Biochemicals, Indianapolis, Indiana) in divalent-free
dilution; gift from Kathy Buckley, Harvard Medical School)43 and FITC- buffer 28 at 37C for 10 min, rinsed and incubated with 1 10 6 M
or TRITC-conjugated anti-mouse IgG (1:1000 dilution; Vector Labs). [3H]glycine (41.1 Ci per mmol; New England Nuclear, Boston, Mass-
achusetts) in oxygenated recording buffer at 37C for 1 h followed by
Isolation and sequencing of chick gephyrin cDNA. The full-length chick rinsing in buffer four times. To determine the total number of counts
gephyrin clone was isolated by screening a White Leghorn E13 chick per CG, single ganglia were dried on Whatman filter paper (2.4 cm)
brain gt10 cDNA library (2 106 pfu) with a [32P]dCTP-labeled rat and counted using a scintillation counter (LS 5000TD, Beckman Coul-
gephyrin cDNA probe (908 bp) using standard procedures44 (chick brain ter, Fullerton, California). To localize [3H]glycine uptake sites, other
cDNA library was provided by Barbara Ranscht, The Burnham Institute, radiolabeled CGs were fixed in 3% glutaraldehyde in PBS (pH 7.4) at
La Jolla, California and the rat gephyrin clone by Heinrich Betz, Max- RT for 1 h or at 4C overnight, and processed for embedding in either
Planck Institute. We isolated a single positive plaque with an insert size of paraffin or Embed 812 (Electron Microscopy Sciences, Fort Washing-
3.1 kb. The identity of the insert as gephyrin was established by Southern ton, Pennsylvania) and for light microscopic autoradiographic analy-
blot hybridization to rat gephyrin cDNA as probe and by sequence analy- sis 48,49. To label synaptic vesicles in presynaptic terminals, freshly
sis (Sequenase Version 2.0 DNA sequencing kit, United States Biochem- dissected CGs were incubated in oxygenated recording buffer with
ical Corporation, Cleveland, Ohio) after subcloning the full-length chick 90 mM KCl and 10 m FM1-43 (Molecular Probes, Eugene, Oregon)
cDNA and smaller restriction enyzme digestion fragments into Blue- at 37C for 1 min 50 , rinsed, then incubated with [ 3 H]glycine and
script II SK vector (Stratagene, La Jolla, California). processed as described above except that alternate sections of the paraf-
fin-embedded ganglia were processed for fluorescence analysis with a
RT-PCR analysis of gephyrin alternative-splice variants. Total RNA was Zeiss Axioskop light microscope.
extracted from CGs (at selected ages ranging from E9 to E15) using Tri
2000 Nature America Inc. http://neurosci.nature.com

Reagent (Molecular Research Center, Cincinnati, Ohio) according to the Glycine release assays. For release assays, E1517 CGs were labeled with
manufacturers instructions. Using an amount of total RNA equivalent [3H]glycine as detailed above and incubated singly at 37C for 15 min in
to that present in a single CG, RT-PCR was done as described45. Seven a 24-well plate in 200 l of recording buffer alone (as a negative control),
different pairs of gephyrin specific primers were used to characterize the 30 mM KCl in recording buffer (to depolarize) or high-K+ buffer con-
alternatively spliced variants expressed in the CG: to flank the C1 and taining either Cd2+ (0.1 mM), low Ca2+ levels (0.2 mM in contrast to
C2 cassettes, we used sense (nt 315334) and antisense (nt 552571)21 5.4 mM), sarcosine (5 mM) or excess cold glycine (1 mM). CGs were
primers; to flank C3, sense (nt 615634 or nt 780797) and antisense pre-incubated in the test condition without high K+ for 10 min. The
(nt 10111028) primers; to flank C4, sense (nt 10111028) and antisense buffer and the CG were counted separately in a scintillation counter (LS
(nt 12041223 or nt 12151232) primers; to flank C3 and C4, sense 5000TD, Beckman).
(nt 615634 or 780797) and antisense (nt 12041223) primers. Ampli-
fication products were separated by 2% agarose-gel electrophoresis and ACKNOWLEDGEMENTS
stained with ethidium bromide. Identities of the products were confirmed We acknowledge Heinrich Betz for providing glycine receptor and gephyrin
by Southern blot hybridization with a chick gephyrin cDNA probe and
antibodies and clones, Yimen Ge (Massachusetts General Hospital, Harvard
sequence analysis.
Medical School) for assistance with the laser-scanning confocal microscope and
Yeast two-hybrid assay. Yeast two-hybrid genetic assay was done as Kathleen Dunlap, Daniel Jay and Tim Turner for advice and comments on the
described46,47. Bait constructs were expressed as recombinant peptide manuscript. This work was supported by NIH grant 21725 to M.H.J.
fragments fused to the DNA-binding protein lexA, using pBTM116 vec-
tor (gift from Peter Pryciak, University of Massachusetts Medical Cen- RECEIVED 9 SEPTEMBER; ACCEPTED 9 DECEMBER 1999
ter). Target constructs were expressed as a fusion peptide with the Gal4
activation domain protein, using the pAD-GAL4 vector (Stratagene). To
test for interactions, chick full-length gephyrin and the major cytoplasmic 1. Zhang, Z. W. & Berg, D. K. Patch-clamp analysis of glycine-induced
loop of either chick GlyR or nAChR 3, 5, 4, 2 or 7 were co- currents in chick ciliary ganglion neurons. J. Physiol. (Lond.) 487, 395405
expressed in the yeast L40 reporter strain as bait and target constructs, (1995).
2. Landmesser, L. & Pilar, G. The onset and development of transmission in the
respectively, and in the reverse orientations in separate experiments. As chick ciliary ganglion. J. Physiol. (Lond.) 222, 691713 (1972).
a negative control, single transformants were tested for transcriptional 3. Landmesser, L. & Pilar, G. Synaptic transmission and cell death during
activation of reporter genes, HIS3 and LacZ. normal ganglionic development. J. Physiol. (Lond.) 241, 737749 (1974).
4. Martin, A. R. & Pilar, G. Dual mode of synaptic transmission in the avian
Electrophysiology. Extracellular recordings were obtained from the intact ciliary ganglion. J. Physiol. (Lond.) 168, 443463 (1963).
5. Martin, A. R. & Pilar, G. Transmission through the ciliary ganglion of the
CG preparation using described techniques with minor modifications27. chick. J. Physiol. (Lond.) 168, 464475 (1963).
Briefly, the E1517 CG was dissected with the pre-and postganglionic 6. Reiner, A. et al. Neurotransmitter organization of the nucleus of Edinger-
nerve stumps attached, mounted on a perfusion recording chamber and Westphal and its projection to the avian ciliary ganglion. Vis. Neurosci. 6,
continuously perfused at about 2 ml per min with oxygenated recording 451472 (1991).
7. Engisch, K. L. & Fischbach, G. D. The development of ACh- and GABA-
buffer. A bipolar tungsten stimulation electrode was used to stimulate activated currents in embryonic chick ciliary ganglion neurons in the absence
the preganglionic nerve stump with current pulses (1020 V, 150 s) at of innervation in vivo. J. Neurosci. 12, 11151125 (1992).
a frequency of 0.1 Hz. A 35 M glass microelectrode filled with nor- 8. Arenella, L. S., Oliva, J. M. & Jacob, M. H. Reduced levels of acetylcholine
mal Krebs solution was used to extracellularly record the field EPSP and receptor expression in chick ciliary ganglion neurons developing in the
compound a.p. triggered by synaptic transmission. The effect of drugs absence of innervation. J. Neurosci. 13, 45254537 (1993).
9. Dale, H. Pharmacology and nerve-endings. Proc. R. Soc. Med. (Lond.) 28,
on the initial slope of the field EPSP and peak-to-peak amplitude of the 319332 (1935).
compound a.p. were measured. Drugs were added directly into the per- 10. Nicoll, R. A. & Malenka, R. C. A tale of two transmitters. Science 281, 360361
fusate. Each trace was averaged 510 times. Glycine, strychnine, hexam- (1998).
ethonium and tetrodotoxin were obtained from Sigma. 11. Jacob, M. H., Lindstrom, J. M. & Berg, D. K. Surface and intracellular
Rough calculations predict that, in the presence of 1 mM exogenous distribution of a putative neuronal nicotinic acetylcholine receptor. J. Cell
Biol. 103, 205214 (1986).
glycine, CG neuron [Na+]i may increase at most by 1.5 mM due to co- 12. Jacob, M. H. Acetylcholine receptor expression in developing chick ciliary
transport via GlyT-1. This change in [Na+]i is not expected to affect the ganglion neurons. J. Neurosci. 11, 17011712 (1991).
driving force for the nAChR current. 13. Williams, B. M. et al. The long internal loop of the alpha 3 subunit targets
nAChRs to subdomains within individual synapses on neurons in vivo. Nat.
Neurosci. 1, 557562 (1998).
Glycine uptake and autoradiographic analysis. For [3H]glycine uptake 14. Jacob, M. H., & Berg, D. K. The ultrastructural localization of -
studies, E1517 CGs were dissected into oxygenated recording buffer, bungarotoxin binding sites in relation to synapses on chick ciliary ganglion
incubated with collagenase type A (1 mg per ml, Boehringer neurons. J. Neurosci. 3, 260271 (1983).

nature neuroscience volume 3 no 2 february 2000 131


2000 Nature America Inc. http://neurosci.nature.com

articles

15. Jacob, M. H., Berg, D. K. & Lindstrom, J. M. Shared antigenic determinant 33. Pow, D. V. Transport is the primary determinant of glycine content in retinal
between the Electrophorus acetylcholine receptor and a synaptic component neurons. J. Neurochem. 70, 26282636 (1998).
on chick ciliary ganglion neurons. Proc. Natl. Acad. Sci. USA 81, 32233227 34. Schwartz, E. A. Depolarization without calcium can release gamma-
(1984). aminobutyric acid from a retinal neuron. Science 238, 350355 (1987).
16. Vernallis, A. B., Conroy, W. G. & Berg, D. K. Neurons assemble acetylcholine 35. Coggan, J. S., Paysan, J., Conroy, W. G. & Berg, D. K. Direct recording of
receptors with as many as three kinds of subunits while maintaining subunit nicotinic responses in presynaptic nerve terminals. J. Neurosci. 17, 57985806
segregation among receptor sites. Neuron 10, 451464 (1993). (1997).
17. Schroder, S., Hoch, W., Becker, C. M., Grenningloh, G. & Betz, H. Mapping of 36. Dumoulin, A. et al. Presence of the vesicular inhibitory amino acid
antigenic epitopes on the alpha 1 subunit of the inhibitory glycine receptor. transporter in GABAergic and glycinergic synaptic terminal boutons. J. Cell
Biochemistry 30, 4247 (1991). Sci. 112, 811823 (1999).
18. Kirsch, J., Wolters, I., Triller, A. & Betz, H. Gephyrin antisense 37. OMalley, D. M., Sandell, J. H. & Masland, R. H. Co-release of acetylcholine
oligonucleotides prevent glycine receptor clustering in spinal neurons. and GABA by the starburst amacrine cells. J. Neurosci. 12, 13941408 (1992).
Nature 366, 745748 (1993). 38. Jo, Y. H. & Schlichter, R. Synaptic corelease of ATP and GABA in cultured
19. Colin, I., Rostaing, P., Augustin, A. & Triller, A. Localization of components spinal neurons. Nat. Neurosci. 2, 241245 (1999).
of glycinergic synapses during rat spinal cord development. J. Comp. Neurol. 39. Smith, M. A., Stollberg, J., Lindstrom, J. M. & Berg, D. K. Characterization of
398, 359372 (1998). a component in chick ciliary ganglia that cross-reacts with monoclonal
20. Feng, G. et al. Dual requirement for gephyrin in glycine receptor clustering antibodies to muscle and electric organ acetylcholine receptor. J. Neurosci. 5,
and molybdoenzyme activity. Science 282, 13211324 (1998). 27262731 (1985).
21. Prior, P. et al. Primary structure and alternative splice variants of gephyrin, a 40. Horch, H. L. & Sargent, P. B. Perisynaptic surface distribution of multiple
putative glycine receptor-tubulin linker protein. Neuron 8, 11611170 classes of nicotinic acetylcholine receptors on neurons in the chicken ciliary
(1992). ganglion. J. Neurosci. 15, 77787795 (1995).
22. Pfeiffer, F., Simler, R., Grenningloh, G. & Betz, H. Monoclonal antibodies and 41. Horch, H. L. & Sargent, P. B. Effects of denervation on acetylcholine receptor
peptide mapping reveal structural similarities between the subunits of the clusters on frog cardiac ganglion neurons as revealed by quantitative laser
glycine receptor of rat spinal cord. Proc. Natl. Acad. Sci. USA 81, 72247227 scanning confocal microscopy. J. Neurosci. 16, 17201729 (1996).
2000 Nature America Inc. http://neurosci.nature.com

(1984). 42. Wang, B. L. & Larsson, L. I. Simultaneous demonstration of multiple


23. Meyer, G., Kirsch, J., Betz, H. & Langosch, D. Identification of a gephyrin antigens by indirect immunofluorescence or immunogold staining. Novel
binding motif on the glycine receptor beta subunit. Neuron 15, 563572 light and electron microscopical double and triple staining method
(1995). employing primary antibodies from the same species. Histochemistry 83,
24. Kins, S., Kuhse, J., Laube, B., Betz, H. & Kirsch, J. Incorporation of a 4756 (1985).
gephyrin-binding motif targets NMDA receptors to gephyrin-rich domains 43. Buckley, K. & Kelly, R. B. Identification of a transmembrane glycoprotein
in HEK 293 cells. Eur. J. Neurosci. 11, 740744 (1999). specific for secretory vesicles of neural and endocrine cells. J. Cell Biol. 100,
25. Zhang, Z. W., Vijayaraghavan, S. & Berg, D. K. Neuronal acetylcholine 12841294 (1985).
receptors that bind alpha-bungarotoxin with high affinity function as ligand- 44. Sambrook, J., Fritsch, E. F. & Maniatis, T. Molecular Cloning: a Laboratory
gated ion channels. Neuron 12, 167177 (1994). Manual (Cold Spring Harbor Press, New York, 1989).
26. Marwitt, R., Pilar, G. & Weakly, J. N. Characterization of two ganglion cell 45. Ikonomov, O. C., Kulesa, M. C., Shisheva, A. C. & Jacob, M. H. Innervation
populations in avian ciliary ganglia. Brain Res. 25, 317334 (1971). and target tissue interactions induce Rab-GDP dissociation inhibitor (GDI)
27. McEachern, A. E., Margiotta, J. F. & Berg, D. K. Gamma-Aminobutyric acid expression during peripheral synapse formation in developing chick ciliary
receptors on chick ciliary ganglion neurons in vivo and in cell culture. ganglion neurons in situ. J. Neurosci. 18, 63316339 (1998).
J. Neurosci. 5, 26902695 (1985). 46. Bartel, P., Chien, C. T., Sternglanz, R. & Fields, S. Elimination of false
28. Sorimachi, M., Rhee, J. S., Shimura, M. & Akaike, N. Mechanisms of GABA- positives that arise in using the two-hybrid system. Biotechniques 14, 920924
and glycine-induced increases of cytosolic Ca2+ concentrations in chick (1993).
embryo ciliary ganglion cells. J. Neurochem. 69, 797805 (1997). 47. Hollenberg, S. M., Sternglanz, R., Cheng, P. F. & Weintraub, H. Identification
29. McGale, E. H., Pye, I. F., Stonier, C., Hutchinson, E. C. & Aber, G. M. Studies of a new family of tissue-specific basic helix-loop-helix proteins with a two-
of the inter-relationship between cerebrospinal fluid and plasma amino acid hybrid system. Mol. Cell Biol. 15, 38133822 (1995).
concentrations in normal individuals. J. Neurochem. 29, 291297 (1977). 48. Jacob, M. H. & Berg, D. K. The distribution of acetylcholine receptors in
30. Attwell, D., Barbour, B. & Szatkowski, M. Nonvesicular release of chick ciliary ganglion neurons following disruption of ganglionic
neurotransmitter. Neuron 11, 401407 (1993). connections. J. Neurosci. 8, 38383849 (1988).
31. Zafra, A. F. et al. Glycine transporters are differentially expressed among CNS 49. Boyd, R. T., Jacob, M. H., McEachern, A. E., Caron, S. & Berg, D. K. Nicotinic
cells. J. Neurosci. 15, 39523969 (1995). acetylcholine receptor mRNA in dorsal root ganglion neurons. J. Neurobiol.
32. Liu, W., Leibach, F. H. & Ganapathy, V. Characterization of the glycine 22, 114 (1991).
transport system GLYT 1 in human placental choriocarcinoma cells (JAR). 50. Ryan, T. A. et al. The kinetics of synaptic vesicle recycling measured at single
Biochim. Biophys. Acta 1194, 176184 (1994). presynaptic boutons. Neuron 11, 713724 (1993).

132 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Enhancement of synaptic
transmission by cyclic AMP
modulation of presynaptic Ih channels
Vahri Beaumont and Robert S. Zucker

Division of Neurobiology, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
Correspondence should be addressed to V.B. (vahri@socrates.berkeley.edu)

Presynaptic activation of adenylyl cyclase and subsequent generation of cAMP represent an


important mechanism in the modulation of synaptic transmission. In many cases, short- to medium-
2000 Nature America Inc. http://neurosci.nature.com

term modulation of synaptic strength by cAMP is due to activation of protein kinase A and
subsequent covalent modification of presynaptic ion channels or synaptic proteins. Here we show
that presynaptic cAMP generation via serotonin receptor activation directly modulated
hyperpolarization-activated cation channels (Ih channels) in axons. This modulation of Ih produced an
increase in synaptic strength that could not be explained solely by depolarization of the presynaptic
membrane. These studies identify a mechanism by which cAMP and Ih regulate synaptic plasticity.

Serotonin is an important neuromodulator of synaptic trans- amplitude (Fig. 1a) with an EC50 of 60 19 nM and a Hill coef-
mission in many phyla17. In crustaceans, serotonin acts as a neu- ficient of 1.14 0.22 (n = 4; Fig. 1d). Simultaneous intracellular
rohormone; on binding to a Gs-coupled receptor8, it strongly recording of axon membrane potential revealed an associated
enhances neuromuscular transmission by increasing the num- depolarization of the resting potential by 10 mV (Fig. 1a and
ber of quanta released per action potential911. Studies on both c) and a slight reduction in action potential amplitude and the
the inhibitor (GABAergic) and excitor (glutamatergic) nerve area beneath its voltage trace, consistent with previous reports8,15.
innervating the muscle have implicated two distinct processes: Forskolin, an activator of adenylyl cyclase, partially mimicked
serotonin increases the absolute number of vesicles available for the effects of serotonin (Fig. 1b and c), maximally increasing EJP
transmitter release12, and serotonin increases the kinetics of amplitude by 120% (EC50, 19 10 M; Hill slope, 1.09 0.26;
release from the inhibitor nerve13. n = 3), and depolarizing the axon membrane by 7 mV. Ampli-
Serotonin alters neither resting presynaptic [Ca2+]i nor cal- tude of the action potential and area beneath its trace were also
cium influx during an action potential in crayfish nerve termi- slightly reduced. The membrane-permeable cAMP analog 8-Br-
nals 13,14 . Its actions seem to be mediated by at least two cAMP (300 M) mimicked the effect of forskolin, enhancing EJP
second-messenger systems, one involving phospholipase C amplitude by 80 12% (n = 6).
(PLC) and another involving adenylyl cyclase8,15. Serotonin We next sought to determine the role of endogenous cAMP
enhancement of transmission is blocked by presynaptic injec- generation in serotonin-induced synaptic enhancement. The phos-
tion of a selective PLC inhibitor8. The downstream messenger phodiesterase inhibitor IBMX prevents breakdown of cAMP, so
and its target that mediate this enhancement are uncertain, but we would expect it to potentiate serotonin enhancement if cAMP
this pathway may involve cAMP. Compounds increasing were generated after serotonin receptor activation. Application of
[cAMP]i can mimic and potentiate serotonin action15,16, and a maximal concentration of serotonin (300 nM) increased EJP
serotonin increases cAMP levels when added to lobster neuro- amplitude by 304 46%. After washout (2 h) to allow EJP ampli-
muscular preparations, although a pre- or postsynaptic locus tude to return to pre-drug levels, subsequent incubation in a low
for cAMP production was undetermined17. In addition, presy- concentration of IBMX (1 M) increased EJP amplitude by
naptic injection of an adenylyl cyclase inhibitor reduces sero- 30 19%. When this EJP amplitude was taken as the new base-
tonin enhancement15. line, application of serotonin in the continued presence of IBMX
Here we show that cAMP generation is an important com- potentiated EJP enhancement (457 122% increase in EJP ampli-
ponent of serotonin enhancement of synaptic transmission, and tude, n = 5, p < 0.1, Fig. 1e). A second application of serotonin in
furthermore, that the target for cAMP is presynaptically locat- the absence of IBMX produced no increase in EJP amplitude over
ed Ih channels. that seen during the first application (p > 0.1, n = 4).
As IBMX is also a nonselective adenosine-receptor antagonist,
RESULTS we investigated whether the observed potentiation might be due
Serotonin-induced enhancement involves cAMP to inhibition of adenosine receptors rather than inhibition of
We recorded excitatory junction potentials (EJPs) from crayfish phosphodiesterase. At a concentration 100-fold higher than that
muscle cells at the neuromuscular junction while stimulating used for IBMX, application of the nonselective adenosine recep-
innervating glutamatergic axons. Superfusion of serotonin (300 tor antagonist, 8-(p-sulfophenyl)-theophylline (8-PST, 100 M;
nM, 25 min) resulted in a maximal increase of 310% in EJP n = 5), similarly increased basal EJP amplitude by 43 16%

nature neuroscience volume 3 no 2 february 2000 133


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 1. Serotonin and forskolin a Control Serotonin (300 nM)


c
enhance synaptic transmission.

Axon depolarization
(a, b) Intracellular recording of
axonal action potentials (APs,
Pre) and muscle excitatory

serotonin

8-BrcAMP
(mV)

forskolin
junction potentials (EJPs; Post)
Post
before (control) and after
application of serotonin (300
nM, 25 min; a) and forskolin Pre
(30 M, 25 min; b). Each trace
is the average of all EJPs/APs
recorded for 1 min at 2-Hz
Control Forskolin (30 M)
b

Percent increase
stimulation. Scale bars, 25 ms

8-BrcAMP
forskolin
EJP amplitude
and 1 mV (Post) or 30 mV

serotonin
(Pre). (c) Bar charts summariz-
ing the axonal depolarization
(top) and enhancement of EJP Post
amplitude (bottom) induced by
serotonin (300 nM), forskolin Pre
(30 M) or 8-Br cAMP (300
2000 Nature America Inc. http://neurosci.nature.com

M); n = 412. (d) Cumulative


concentrationresponse curves
for serotonin () and forskolin
(). The lower maximal d e
increase in EJP amplitude with
IBMX (1 M)
forskolin than with serotonin
Fold increase in EJP amplitude

demonstrates that adenylyl serotonin (300 nM)


% increase control EJP

cyclase activation cannot


entirely account for serotonin-
induced enhancement. Each
amplitude

point represents the


mean s.e. of four experi-
ments. (e) Time course of
serotonin (300 nM)-induced
enhancement of EJP amplitude
(). After EJPs returned to
control amplitudes following
washout of serotonin, each
log10 [drug] (log M) Time (min)
preparation was incubated for
25 min with a low concentra-
tion of the phosphodiesterase inhibitor IBMX (1 M). IBMX alone resulted in a small enhancement of EJP amplitude (normalized to control in graph). A sec-
ond serotonin application was potentiated in the presence of IBMX (n = 5, ). This effect might be explained by increased cAMP generation over control
levels in response to serotonin application.

(IBMX response, 30 19%, n = 5). However, in contrast to IBMX, (Fig. 2b; p < 0.05, Students t-test). Forskolin significantly
8-PST did not potentiate the serotonin (300 nM) response; the increased frequency in 3 of 6 experiments (Kolmogorov-Smirnov
increase in EJP amplitude induced by serotonin in the presence test, p < 0.05) with a mean increase in frequency of 42 33% and
of 8-PST was not different from that elicited by serotonin in the no increase in amplitude of mEJPs (p > 0.1, Students t-test,
absence of 8-PST (3 11% difference; p > 0.05, n = 5). Therefore, Fig. 2b). Consistent with serotonins enhancement of the size of
IBMX potentiation of the serotonin-induced enhancement seems the vesicle pool12, this finding supports the assumption that the
to result from phosphodiesterase inhibition, although the effect locus of action of both serotonin and forskolin is presynaptic.
of IBMX alone on EJP amplitude may be due to a nonspecific
action, or even to inhibition of adenosine receptors. PKA is not involved
The downstream effects of cAMP at the crayfish neuromuscu-
Serotonin and forskolin act presynaptically lar junction have previously been attributed to activation of
The locus of action of both serotonin and forskolin (and hence PKA 15 . We tested the effects of two membrane-permeable
cAMP generation) was examined using analysis of miniature EJPs inhibitors of PKA on the enhancement induced by serotonin
(mEJPs). In Normal Van Harrevalds solution containing TTX and 8-Br-cAMP. A submaximal concentration of serotonin (100
(1 M), the mean frequency of mEJPs recorded over a 30-minute nM, 25 min) elicited an increase in EJP amplitude of 144 24%.
period was 0.31 0.1 Hz with a mean amplitude of 254 36 V In the presence of 30 M H-7, which should fully inhibit PKA,
(n = 12). After incubation for 20 minutes in either serotonin PKC and PKG (Ki = 36 M18,19), a second application of sero-
(1 M) or forskolin (30 M), the same muscle fiber was record- tonin resulted in an amplitude increase no different from the
ed for 30 minutes in the presence of the drug. In 5 of 6 paired control response (148 33%, n = 4, p > 0.05). H-7 was simi-
experiments, serotonin induced a significant increase in frequency larly ineffective against the increase induced by 8-Br-cAMP (8-
(p < 0.05, Kolmogorov-Smirnov test) but not amplitude of mEJPs Br-cAMP alone, 74 23%; 8-Br-cAMP plus H-7, 95 38%;
(Fig. 2a and b), with a mean increase in frequency of 64 11% n = 3; p > 0.05). However, H-7 (30 M) prevents an unrelated

134 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

presynaptic phorbol ester-induced potentiation of transmis- shifts of the activation curve to more depolarized potentials2025
sion at the crayfish neuromuscular junction (V.B. and R.S.Z., would explain depolarization on addition of cAMP analogs8,15.
unpublished observation), demonstrating that this compound The presence of Ih channels in the crayfish excitor axon was
permeates the axon sufficiently to inhibit PKC. investigated by intracellular injection of hyperpolarizing current
In addition to H-7, we tested the highly specific, cell-perme- pulses (500 ms, 5 to 50 nA) into the axon. Upon hyperpolar-
able PKA inhibitor Rp-8-Br-cAMPS. Incubation with Rp-8-Br- ization of the membrane beyond resting potential, Ih should
cAMPS (300 M) alone caused a 40 13% increase in EJP become activated, typically producing a depolarization sag back
amplitude, suggesting that this cAMP analog may mimic cAMP toward the resting potential, whereas termination of the pulse
itself. When serotonin was also added, EJP amplitude increased by results in a transient depolarizing overshoot of the original resting
292 68% of pre-drug amplitude, not significantly different from potential (after-depolarization potential; ADP)25,26. ADPs were
the paired response with serotonin alone (324 125%, n = 3; produced by injecting current pulses of different amplitudes
Fig. 3a). Similar effects of Rp-8-Br-cAMPS were found against (Fig. 4). With large current injections, the ADP was of sufficient
8-Br-cAMP-induced increases in EJP amplitude (Fig. 3b). Because amplitude to initiate firing of action potentials, as demonstrated
Rp-8-Br-cAMPS is a cAMP analog, and it increased EJP ampli- for Ih channels in many neurons25. When the Ih blocker Cs+ (1
tude significantly, these results suggest that the target of cAMP mM) was applied to the preparation, resting membrane poten-
may be a cyclic nucleotide-binding effector other than PKA. tial hyperpolarized by approximately 4 mV, and ADP amplitude
was significantly reduced (Fig. 4a). Injection of between 5 and
Presynaptic Ih channels are modulated by cAMP 50 nA of current elicited ADPs (Fig. 4b; n = 6) that were blocked
2000 Nature America Inc. http://neurosci.nature.com

Depolarization of the axon by forskolin, 8-Br-cAMP and serotonin by Cs+ (n = 3) or the specific Ih blocker ZD7288 (ref. 27; n = 3).
(Fig. 1ac) suggests that presynaptic Ih channels may be targets for These data not only demonstrate the presence of Ih channels in
cAMP. Indeed, modulation of Ih channels by the direct binding of excitor axons, but also suggest that these channels contribute to
cyclic nucleotides (including Rp-8-Br-cAMPS20) and subsequent the axons resting potential. However, it should be noted that,
a
Control

Serotonin (1 M)

b
Fig. 2. Serotonin and forskolin act
at a presynaptic locus to enhance
synaptic transmission. (a) 20-s
sample traces of miniature EJPs,
Cumulative percent

measured in NVH solution con-


taining TTX (1 M) before (con-
trol) and after a 20-min incubation
in serotonin (1 M). Scale bars,
0.5s, 0.3 mV. (b) Left, cumulative
probability plot for inter-event Control 100 V
interval after analysis of mEJPs
Serotonin (1 M)
recorded for 30 min in NVH solu-
20 ms
tion alone (____) or in the presence
of serotonin (----) indicates signifi-
cantly higher event frequency in Inter-event interval (s)
serotonin (0.57 Hz versus 0.33 Hz,
control). Right, superimposed
average mEJPs with and without
serotonin show similar amplitudes.
(c) Percent change in both mean c Percent change in mean frequency Percent change in mean amplitude
frequency (left) and mean ampli-
tude (right) of mEJPs after applica-
tion of either forskolin (open bars)
or serotonin (filled bars) Each bar
represents the mean s.e. of six Forskolin
experiments. Both forskolin and
serotonin application increased Serotonin
mEJP frequency without signifi-
cantly altering amplitude.

nature neuroscience volume 3 no 2 february 2000 135


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 3. Rp-8Br-cAMPS elevates


EJP amplitude and does not sig- a b
nificantly block serotonin Rp-8-Br-cAMPS Rp-8-Br-cAMPS

Percent increase EJP amplitude

Percent increase EJP amplitude


enhancement of transmission. serotonin 8-Br-cAMPS
(a) Time course of the increase
of EJP amplitude in response to
serotonin (100 nM; ). After
serotonin was washed out and
EJP amplitude reached a steady
baseline, preparations were incu-
bated with the membrane-per-
meable specific PKA inhibitor
Rp-8Br-cAMPS (300 M).
Surprisingly, this incubation
enhanced EJP amplitude (,*) by
approximately 50% over control. Time (min) Time (min)
Serotonin-induced enhancement
was affected little by the PKA inhibitor (). (b) Time course of the increase of EJP amplitude by the membrane-permeable cAMP analog 8-Br-cAMP
(, 300 M). After washout of 8-Br-cAMP, EJP amplitude reached a steady baseline, and preparations were incubated with Rp-8Br-cAMPS (300 M).
Again, this incubation increased EJP amplitude 48 4% over control (,*) Addition of 8-Br-cAMP together with Rp-8Br-cAMPS resulted in a slightly
2000 Nature America Inc. http://neurosci.nature.com

smaller increase in EJP amplitude (78 7%; ) than 8-Br-cAMP applied alone (120 30%, ).

whereas ADPs elicited by current injection up to and including effects on the action potential seen with Cs+, but did not affect
30 nA could be blocked effectively, ADPs elicited by larger current serotonin-induced depolarization (Fig. 5a). ZD7288
injections were not completely blocked by either Cs+ or ZD7288 (100 nM100 M; 25 min) potently reduced serotonin-induced
(Fig. 4a and b), suggesting that ADPs evoked by injection of more depolarization of the axon, with an IC50 similar to that for its
than 30 nA may involve conductances other than Ih channels. action on I h channels 27,28 (IC 50, 6 2 M; Hill coefficient,
To determine whether Ih channels were modulated by cAMP, 1.19 0.01; n = 4, Fig. 5b).
we attempted to block the serotonin- and forskolin-induced depo- Serotonin seemed to alter Ih activity through cAMP genera-
larization of the axon (Fig. 1a and b) using Cs+ and ZD7288. tion, as application of forskolin (30 M) also produced axon
Application of serotonin (300 nM) resulted in a maximum axon depolarization of 6.3 0.6 mV from a mean resting potential of
depolarization of 9.5 0.75 mV from a mean resting membrane 71 1.4 mV (n = 8, Fig. 5c and d). The significantly smaller size
potential of 71 1 mV (n = 12; Fig. 5a) within 16 minutes of of the depolarization with forskolin than with serotonin
serotonin application. Addition of Cs+ (3 M3 mM) reversed (10 mV) probably resulted from the barely submaximal con-
this depolarization in a concentration-dependent manner (IC50, centration of forskolin used (Fig. 1d). Incubation with Cs +
260 20 M; Hill coefficient, 1.26 0.05; n = 5, Fig. 5b). A slight (1 mM, n = 4) or ZD7288 (30 M, n = 4) abolished the depolar-
broadening of the action potential and an increase in area beneath ization induced by a second application of forskolin (Fig. 5c and
the voltage trace was observed in Cs+, consistent with additional d). In summary, we demonstrated that cAMP generated in the
block of a potassium conductance. However, addition of the potas- axon acts on Ih, resulting in depolarization with a time course
sium-channel blocker Ba2+ (1 mM) to the nerve reproduced the paralleling that of synaptic enhancement.

a b
20 nA 40 nA
Iinj
Peak amplitude ADP (mV)

RMPaxon

Hyperpolarizing pulse (nA)

Fig. 4. Ih channels are involved in maintenance of resting membrane potential, and Ih activation results in anomolous membrane rectification in exci-
tatory axons. (a) Injection of axons with hyperpolarizing current (500 ms) resulted in after-depolarizing potentials (ADPs; traces in black). Resting
membrane potential of the axon (dashed line) was 66 mV. Membrane responses during the pulse were truncated. With large hyperpolarizing pulses
(typically 40 nA), the ADP was sufficient to reach threshold for action potential generation (APs truncated in trace). Scale bar, 2 s, 10 mV. In the
same axon, resting membrane potential became relatively hyperpolarized in the Ih channel blocker Cs+ (1 mM; compare gray traces with dashed line),
and ADP amplitudes in response to hyperpolarizing pulses were reduced (gray traces). (b) Plots of peak ADP amplitude versus size of injected hyper-
polarizing current in controls (, n = 6), with Cs2+ (, 1 mM, n = 3) or with ZD7288 (, 30 M, n = 3).

136 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Modulation of Ih by cAMP results in synaptic enhancement blocked in the presence of ZD7288 (30 M), which decreased
To investigate the involvement of Ih modulation in synaptic synaptic enhancement by 67 6% (Fig. 6c) and 70 18%
enhancement, we studied the increase of EJP amplitude by sero- (Fig. 6d) relative to the respective control responses. Enhance-
tonin, forskolin and 8-Br-cAMP in the absence and presence of ment of EJP amplitude by 8-Br-cAMP was blocked by Cs+ equal-
ZD7288 (30 M) or Cs+ (1 mM), with Ba2+ (1 mM) as a control. ly well (82 2% reduction of the control response, n = 3).
In the absence of any other drug, serotonin (100 nM) caused However, Cs+ was less effective in blocking forskolin enhance-
a 2.9 0.3-fold increase in EJP amplitude (control response; ment, reducing the response to only 31 15% of the control
n = 13). Two hours later, recordings from the same muscle cells response (n = 3). Although the reason for this is not obvious,
incubated in Cs+ (1 mM) 10 minutes before and during a second Cs+ blockade of a potassium conductance in these cells may have
serotonin application showed a 43 7% reduction compared with induced a depolarization that counteracted the effect of block-
control responses to serotonin (1.8 0.2-fold increase in EJP ampli- ing Ih to a greater extent than observed in other experiments.
tude; p < 0.05, n = 6), whereas application of 1 mM Ba2+ before It is conceivable that the reduction of the serotonin and
and during a third serotonin application insignificantly increased forskolin response by ZD7288 and Cs+ ions may have been a
EJP amplitude 17 13% over controls, consistent with the slight result only of the membrane hyperpolarization produced by
widening of the action potential we observed (n = 6; Fig. 6a and these agents, rather than by a specific block of an Ih conduc-
b). A 25-minute application of ZD7288 before and during sero- tance. We felt this was unlikely, as hyperpolarizing the axon
tonin application was as effective as Cs+ in reducing synaptic by 5 mV by manipulating extracellular potassium has no
enhancement, decreasing the response to serotonin by 42 13% effect on either serotonin-induced axonal depolarization or
2000 Nature America Inc. http://neurosci.nature.com

from the control (n = 7, p < 0.05; Fig. 6a). Neither Cs+ nor ZD7288 serotonin-induced synaptic enhancement14,29. Nevertheless,
applied on its own reduced basal EJP amplitude (n = 4 each). we eliminated hyperpolarization of the axon in the presence
Both forskolins (30 M) and 8-Br-cAMPs (300 M) of ZD7288 (30 M) by slightly elevating extracellular potas-
enhancement of EJP amplitude (1.4 0.2-fold increase, n = 9 sium concentration to 6.757.0 mM (Fig. 7a). Even with the
and 0.8 0.1-fold increase, n = 6, respectively) were markedly offset of hyperpolarization afforded by increased [K + ] e ,

a c Cs+ (1 mM)
Cs+ Ba2+ forskolin forskolin
serotonin serotonin serotonin
RMP axon(mV)

RMP axon(mV)

Time (min) Time (min)

d ZD7288 (30 M)
b
forskolin forskolin
Depolarization of axon

RMP axon(mV)
(mV)

log10 [Ih inhibitor] (log M) Time (min)

Fig. 5. Ih modulation by cAMP generation results in axon depolarization. (a) Application of serotonin (300 nM) to axons resulted in an approximately
10-mV membrane depolarization (n = 5, ). After washout of serotonin, membrane depolarization slowly reversed to pre-serotonin levels.
Application of Cs+ (1 mM) to the axon resulted in a rapid membrane hyperpolarization of approximately 5 mV. Subsequent application of serotonin
in the presence of Cs+ was ineffective in eliciting a membrane depolarization. After washout of Cs+, incubation with Ba2+ (1 mM) affected neither
resting membrane potential nor subsequent depolarization in response to serotonin (n = 4; typical s.e., 1.5 mV). (b) Cumulative concentrationinhi-
bition curves for Cs+ (; n = 3 separate experiments) and the irreversible Ih blocker ZD7288 (; n = 4) against axon depolarization induced by sero-
tonin (300 nM). Each point represents the mean s.e. (c, d) Forskolin (30 M) mimics serotonin at the axon, suggesting that cAMP generation is
responsible for increased Ih activation and resulting depolarization. The effects of forskolin can be blocked by either Cs2+ (c; 1 mM) or ZD7288
(d; 30 M).

nature neuroscience volume 3 no 2 february 2000 137


2000 Nature America Inc. http://neurosci.nature.com

articles

forskolin-induced synaptic enhancement after ZD7288 treat- several hundred microns of boutons innervating a muscle fiber,
ment was reduced to the same extent as without elevating which was also impaled with a microelectrode. We then simulta-
[K+]e (68 23% reduction, n = 3, Fig. 7b). neously recorded both evoked action potentials (2 Hz) in the exci-
In summary, pooled results from the above experiments show tor nerve and subsequent EJPs in the muscle (Fig. 8a). After 10
that Ih activation by cAMP was responsible for 62 7% (n = 18) minutes, stimulation was stopped and the axon hyperpolarized
of the cAMP-induced synaptic enhancement, and activation of Ih by injection of 30 nA for 1 minute via the intracellular electrode,
via cAMP generation accounted for 43 6% (n = 13) of the sero- a protocol that elicits a large ADP, but is not sufficient to induce
tonin-induced enhancement of synaptic strength. spontaneous firing of action potentials (Fig. 4). The increased
time of current injection used in this experiment (1 min versus
Axonal hyperpolarization enhances synaptic transmission 0.5 s in Fig. 4) did not significantly affect the amplitude of the
Our results thus far implicated cAMP modulation of Ih channels ADP (13 6 mV after 1-min and 16 4 mV after 0.5-s current
as a vital component of the increase in synaptic strength observed injection) or its sensitivity to ZD7288 (79 21% block and 87
on addition of either serotonin or forskolin. We extended this 18% block, respectively; n = 3), suggesting that this prolonged
observation to see whether voltage-gated activation of Ih channels current protocol elicited an ADP indicative mainly of Ih channel
per se was sufficient to increase synaptic transmission at the cray- activation. During axon hyperpolarization, we often noted an
fish neuromuscular junction. First, the fluorescent dye Fura-2 (17 increase in the number of spontaneous EJPs (Fig. 8a) which was
mM in 200 mM KCl) was iontophoresed (10 nA, 10 min) into not observed previously using methods that polarize large axon-
the axon via a microelectrode. Diffusion of the dye along the al branches30. If no increase in spontaneous events was detected
2000 Nature America Inc. http://neurosci.nature.com

length of the axon allowed visualization of small tertiary branch- during hyperpolarization, it was assumed that the hyperpolariz-
es and boutons innervating individual muscle fibers. Using a new ing electrode was too far from boutons to elicit a response in the
intracellular electrode (3 M KCl), we reimpaled the axon within recorded muscle fiber, and the experiment was discounted (4 of

a Percent of control 5HT increase


b
1 mM Cs+/Ba2+
Percent increase EJP amplitude

serotonin

control 5HT enhancement

1 mM Ba2+

1 mM Cs + Fig. 6. Inhibition of Ih channels


resulted in a reduction of
ZD7288 (30 M) synaptic enhancement by sero-
c Time (min)
tonin, forskolin and 8-Br-cAMP.
(a) Effects of Ba2+ (1 mM) and
forskolin (30 M)
the Ih blockers Cs+ (1 mM) or
Percent increase EJP amplitude

Percent of control forskolin increase


ZD7288 (30 M) on EJP ampli-
tude enhancement by sero-
tonin (100 nM; upper),
forskolin (30 M; middle, gray
forskolin, control bars) or 8-Br cAMP (300 M;
lower) To allow visual compari-
son of the percent inhibition of
1 mM Ba2+
synaptic enhancement by either
Cs+ or ZD7288, enhancements
1 mM Cs + with serotonin, forskolin or 8-
Br-cAMP alone (control
enhancement) were normal-
ZD7288 d Time (min) ized to 100%. (bd) Data from
which bar charts were gener-
Percent of control 8-Br-cAMP 8-Br-cAMP (300 M) ated. (b) Time course of the
Percent increase EJP amplitude

increase
percent increase in EJP ampli-
8-Br-cAMP, control tude with serotonin alone (100
nM; ) or after concurrent
incubation with Cs+ (1 mM; )
1 mM Ba2+
or Ba2+ (1 mM, _____).
(c, d) Percent increase in EJP
Cs + amplitude during application of
forskolin (c; ) or 8-Br-cAMP
(d; ) alone or after a 30-min
ZD7288 pre-incubation with ZD7288
(30 M, ). Each point repre-
sents the mean s.e. of 47 dif-
Time (min) ferent experiments.

138 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a b sistent with the notion that EJP


enhancement is a result of hyperpo-

Percent increase in EJP amplitude


forskolin larization per se. Indeed, if hyperpo-
KCI 5.4 mM 6.5 mM 7 mM 5.4 mM
larization independent of I h
ZD7288 activation mediated the enhance-
RMPaxon (mV)

ment of EJP amplitude, one would


expect a greater increase after
ZD7288 application, where blockade
of Ih conductance would increase the
hyperpolarization induced by cur-
rent injection. We therefore conclude
that a relatively brief Ih channel acti-
vation, independent of cAMP mod-
Time (min) Time (min) ulation, is also capable of eliciting a
Fig. 7. Reversal of the ZD7288-mediated axonal hyperpolarization does not affect ZD7288 block of more prolonged synaptic enhance-
forskolin-induced synaptic enhancement. (a) The axon was impaled, and ZD7288 (30 M) was applied ment.
after determining the postsynaptic control response to forskolin (b, ). Resulting presynaptic membrane
hyperpolarization was then offset by stepwise increases in extracellular potassium to return the axon to DISCUSSION
2000 Nature America Inc. http://neurosci.nature.com

its original resting potential. The recovery of hyperpolarization when the [K+]e was returned to normal
Neuronal Ih channels subserve fun-
(5.4 mM) demonstrates the irreversibility of Ih block by ZD7288. (b) After determining the postsynaptic
control response to forskolin (), ZD7288 was applied and the resulting hyperpolarization of the axon
damental physiological functions
membrane potential offset back to the original resting potential (see a) before recording the response to such as contribution to membrane
forskolin from a different muscle fiber in the same preparation (, n = 3). ZD7288 still reduced forskolin- potential3134, integration of synaptic
induced synaptic enhancement under these conditions. input to neurons 35 and the rhyth-
micity of various brain regions3639.
Binding of cAMP to Ih results in a
concentration-dependent shift of the
12). After axon hyperpolarization, action potentials and subse- activation curve to more depolarized voltages through a phos-
quent EJPs were again evoked at two Hz. EJP amplitude signifi- phorylation-independent allosteric interaction with the chan-
cantly increased following hyperpolarization, increasing an average nel40,41. Thus receptors both positively and negatively coupled to
of 54 11% by 1020 minutes after current injection (Fig. 8a and adenylyl cyclase can shift Ih activation by up to 20 mV25, and
b; p < 0.05, n = 8). -adrenoceptors23, serotonin receptors23,42,43, A1 adenosine recep-
In separate experiments, ZD7288 (30 M) was applied for tors44 and -opioid receptors21 regulate Ih in this way. The chan-
30 minutes before and during the 30 nA
hyperpolarization of the axon (Fig. 8b). a Before hyperpolarization Before hyperpolarization 10 min after hyperpolarization
Again, individual experiments were aban-
doned if spontaneous release was not seen During hyperpolarization
during the hyperpolarizing pulse, although Post Post
this was rare (1 of 7) in the presence of
ZD7288. The increase in spontaneous Pre Pre
release observed during hyperpolarization 1 mV
with ZD7288 probably indicated that hyper- 0.5 s
polarizing current pulses applied during Ih
block resulted in greater changes in poten-
tial compared with those in control trials. b
EJP amplitude sometimes increased slight- Hyperpolarization
(30 nA, 1 min)
ly (average 11 8%) and transiently
Percent increase EJP

between 1 and 5 minutes after the termina-


amplitude

tion of the pulse, correlating positively with


the level of spontaneous activity during the
hyperpolarization, although this was not
quantified further. However, ZD7288 com-
pletely prevented the enhancement of EJP
amplitude between 10 and 20 minutes after
current injection (2 5% reduction from
basal amplitude, n = 6, significantly different Time (min)
from the control trials; p < 0.001, Students
Fig. 8. Brief activation of Ih channels by hyperpolarization is sufficient to induce long-lasting
unpaired t-test).
enhancement of EJP amplitude. (a) Intracellular recording of axonal action potentials (APs; pre)
It should be noted that, although this and muscle excitatory junction potentials (EJPs; post) before (control) and 10 minutes after
experiment is unavoidably compromised by hyperpolarizing current injection (1 min, 30 nA) into the axon to activate I channels. Each trace
h
the probable difference in axon membrane is the average of all EJPs/APs recorded for 1 min at 2-Hz stimulation. Scale bars, 25 ms and 1 mV
potential change elicited by the current pulse (post) or 25 mV (pre). Spontaneous EJPs increase upon hyperpolarization (middle panel).
with or without Ih channel block, the pre- (b) Time course of EJP amplitude before, during and after hyperpolarization (30 nA, 1 min) to
vention of any increase by ZD7288 is incon- activate presynaptic Ih channels with ZD7288 (; 30 M; n = 6) or without ZD7288 (; n = 8).

nature neuroscience volume 3 no 2 february 2000 139


2000 Nature America Inc. http://neurosci.nature.com

articles

nels we describe were similar to other reported Ih channels based City, California). EJP amplitudes were measured off-line (Clampfit 6.05,
on their activation by hyperpolarization, the contribution made Axon Instruments). For miniature EJP recording, beveled sharp electrodes
to axonal resting potential, their increased activation by cAMP (resistance 510 M) were used to continuously acquire recordings sub-
and their selective pharmacological block by both Cs + and sequently filtered at 1 kHz, digitized at 2.5 kHz and analyzed off-line using
ZD7288. The Ih current is a mixed cation current (Na+/K+)25, and Minianalysis Program version 4.0.1 (Synaptosoft, Leonia, New Jersey).
under physiological conditions, current is carried predominant-
Data presentation and statistical analysis. As control EJP amplitudes were
ly by the movement of sodium ions. In the crayfish axon, this extremely variable from fiber to fiber, results were expressed as percent
also seems to be the case, as serotonin-induced depolarization of change from control EJP amplitude, taken as the average EJP amplitude
membrane potential was prevented when external sodium was over ten minutes of continuous recording in the absence of drug. Data
replaced with choline15. are plotted as mean s.e. percent change from this control level. When
The mechanism by which serotonin-induced activation of effects of different drugs were tested within a single muscle fiber, results
Ih channels in the axon results in an increase in the total num- were analyzed by Students paired t-test; the Kolmogorov-Smirnov test
ber of vesicles available for release12 remains elusive. It is clear was used to compare differences in cumulative probability for analysis of
from previous studies that the amplitude of depolarization per mEJPs. Significance was assumed if p < 0.05, unless otherwise stated.
se elicited by serotonin or forskolin is insufficient to account
for the enhancement of neurotransmission, as depolarizing the ACKNOWLEDGEMENTS
axon by the same amount using either high [K+]14,29 or current We thank Russell English for technical assistance. This work was supported by
injection through intracellular recording electrodes45 could not
2000 Nature America Inc. http://neurosci.nature.com

NSF Grant IBN-9722826.


mimic the increase in synaptic strength. In addition, the obser-
vation that EJP amplitude remains elevated for up to 20 min-
utes after a relatively brief (1 minute) axonal hyperpolarization RECEIVED 24 AUGUST; ACCEPTED 30 NOVEMBER 1999
would suggest that Ih activation is required only for initiating
a downstream process resulting in synaptic enhancement.
1. Sugita, S., Goldsmith, J. R., Baxter, D. A. & Byrne, J. H. Involvement of pro-
Synaptic enhancement after axon hyperpolarization is observed tein kinase C in serotonin-induced spike broadening and synaptic facilitation
not only in crayfish30 but also in vertebrate junctions46. Before in sensorimotor connections of Aplysia. J. Neurophysiol. 68, 643651 (1992).
knowledge of Ih channels, either a prolongation of the action 2. Hori, Y., Endo, K. & Takahashi, T. Long-lasting synaptic facilitation induced
by serotonin in superficial dorsal horn neurones of the rat spinal cord.
potential30 or a hyperpolarization-dependent mobilization of J. Physiol. (Lond.) 492, 867876 (1996).
vesicles was assumed responsible30,47. As we did not stimulate 3. Sun, Z. Y. & Schacher, S. J. Binding of serotonin to receptors at multiple sites
the nerve and record EJPs during hyperpolarization, and action is required for structural plasticity accompanying long-term facilitation of
Aplysia sensorimotor synapses. J. Neurosci. 18, 39914000 (1998).
potentials were unaltered after hyperpolarization, the former 4. Schuman, E. M. & Clark, G. A. Synaptic facilitation at connections of
hypothesis can be rejected. However, active Ih-dependent mobi- Hermissenda type B photoreceptors. J. Neurosci. 14, 16131622 (1994).
lization of vesicles from non-releasable to releasable pools rep- 5. Stubli, U. & Otaky, N. Serotonin controls the magnitude of LTP induced by
theta bursts via an action on NMDA-receptor-mediated responses. Brain Res.
resents an attractive possibility, and the role of microtubule and 643, 1016 (1994).
actin transport pathways in this cAMP-induced enhancement 6. Mintz, I. & Korn, H. Serotonergic facilitation of quantal release at central
are being investigated. inhibitory synapses. J. Neurosci. 11, 33593370 (1991).
7. Mitoma, H., Kobayashi, T., Song, S. Y. & Konishi, S. Enhancement by serotonin
Ih clearly can be included with other presynaptic voltage- and of GABA-mediated inhibitory synaptic currents in rat cerebellar Purkinje cells.
ligand-gated ion channels modulated by neurotransmitters to Neurosci. Lett. 173, 127130 (1994).
inhibit or facilitate transmission. The many reports of Ih chan- 8. Dixon, D. & Atwood, H. L. Phosphatidylinositol systems role in serotonin-
induced facilitation at the crayfish neuromuscular junction. J. Neurophysiol. 62,
nels in vertebrate and invertebrate axons42,4749 suggest physio- 239246 (1989).
logical importance of cAMP-induced Ih modulation in regulating 9. Dudel, J. Facilitatory effects of 5-hydroxy-tryptamine on the crayfish neuromuscular
synaptic strength in a variety of organisms. junction. Naunyn Schmiedebergs Arch. Pharmacol. 249, 515528 (1965).
10. Fischer, L. & Florey, E. Modulation of synaptic transmission and excitation-
contraction coupling in the opener muscle of the crayfish, Astacus leptodactylus,
METHODS by 5-hydroxytryptamine and octopamine. J. Exp. Biol. 102, 187198 (1983).
Crayfish (Procambarus clarkii, 23 inches) were obtained from Niles Bio- 11. Glusman, S. & Kravitz, E. A. The action of serotonin on excitatory nerve
stimulation in lobster nerve-muscle preparations. J. Physiol. (Lond.) 325,
logical (Sacramento, California). Preparation of the innervated dactyl 223241 (1982).
opener muscle of the first walking leg is described14. Autotomized legs 12. Wang, C. & Zucker, R. S. Regulation of synaptic vesicle recycling by calcium and
were continuously superfused by a gravity-fed perfusion system with serotonin. Neuron 21, 155167 (1998).
Normal Van Harrevalds solution, containing 195 mM NaCl, 13.5 mM 13. Vyshedskiy, A., Delaney, K. & Lin, J.-W. Neuromodulators enhance transmitter
CaCl2, 5.4 mM KCl, 2.6 mM MgCl2 and 10 mM Na-HEPES at pH 7.4 release by two separate mechanisms at the inhibitor of crayfish opener muscle.
J. Neurosci. 18, 51605169 (1998).
and 1417C. Serotonin, H-7, 8-(phenylsulfonyl)-theophylline and 8- 14. Delaney, K., Tank, D. W. & Zucker R. S. Presynaptic calcium and serotonin-
Br-cAMP were obtained from Sigma; Fura-2 from Molecular Probes mediated enhancement of transmitter release at crayfish neuromuscular
(Eugene, Oregon); and ZD7288 from Tocris Cookson (Ballwin, Mis- junction. J. Neurosci. 11, 26312643 (1991).
souri). All other drugs were from Calbiochem (La Jolla, California). All 15. Dixon, D. & Atwood, H. L. Conjoint action of phosphatidylinositol and
paired control experiments contained final concentrations of DMSO adenylate cyclase systems in serotonin-induced facilitation at the crayfish
neuromuscular junction. J. Neurophysiol. 62, 12511259 (1989).
equal to those used for drug delivery. 16. Enyeart, J. Cyclic AMP, 5-HT, and the modulation of transmitter release at the
crayfish neuromuscular junction. J. Neurobiol. 12, 505513 (1981).
Electrophysiology. Sharp electrodes were used to impale both proximal 17. Goy, M. F. & Kravitz, E. A. Cyclic AMP only partially mediates the actions of
serotonin at lobster neuromuscular junctions. J. Neurosci. 9, 369379 (1989).
muscle fibers (electrode resistance, 1225 M) and/or either primary or 18. Boulis, N. M. & Davis M. Blockade of the spinal excitatory effect of cAMP on the
secondary branches of the excitor nerve axon (beveled electrode resis- startle reflex by intrathecal administration of the isoquinoline sulfonamide H8:
tance, 2545 M). Excitor nerves were stimulated (2 Hz) during record- comparison to the protein kinase C inhibitor H7. Brain Res. 525, 198204 (1990).
ing using a suction electrode containing the excitor axon freed from the 19. Quick, J., Ware, J. A. & Driedger P. E. The structure and biological activities of
meropodite segment of the leg. Signals were amplified (Neuroprobe 1600 the widely used protein kinase inhibitor, H7, differ depending on the
commercial source. Biochem. Biophys. Res. Commun. 187, 657663 (1992).
Amplifier, A-M systems Carlsborg, Washington), filtered at 2 kHz, and 20. Ingram, S. L. & Williams, J. T. Modulation of the hyperpolarization-activated
digitized at 5 kHz and the average of all EJPs elicited each minute was current (Ih) by cyclic nucleotides in guinea pig primary afferent neurones.
saved to computer using pClamp7 software (Axon Instruments Foster J. Physiol. (Lond.) 492, 97106 (1996).

140 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

21. Ingram, S. L & Williams, J. T. Opioid inhibition of Ih via adenylyl cyclase. Neuron 36. Bal, T. & McCormick, D. A. What stops synchronized thalamocortical
13, 179186 (1994). oscillations? Neuron 17, 297308 (1996).
22. Tokimasa, T. & Akasu, T. Cyclic AMP regulates an inward rectifying sodium- 37. Leresche, N., Lightowler, S., Soltesz, I., Jassik-Gerschenfeld, D. & Crunelli, V.
potassium current in dissociated bull-frog sympathetic neurones. J. Physiol. Low-frequency oscillatory activities intrinsic to rat and cat thalamocortical
(Lond.) 420, 409429 (1990). cells. J. Physiol. (Lond.) 441, 155174 (1991).
23. Pape, H.-C. & McCormick, D. A. Noradrenaline and serotonin selectively 38. Dekin, M. S. Inward rectification and its effects on the repetitive firing
modulate thalamic burst firing by enhancing a hyperpolarization-activated properties of bulbospinal neurons located in the ventral part of the nucleus
cation current. Nature 340, 715718 (1989). solitarius. J. Neurophysiol. 70, 590601 (1993).
24. Di Francesco, D. & Mangoni, M. Modulation of single hyperpolarization- 39. Golowasch, J. & Marder, E. Ionic currents of the lateral pyloric neuron of the
activated channels (if) by cAMP in the rabbit sinoatrial node. J. Physiol. (Lond.) somatogastric ganglion of the crab. J. Neurophysiol. 67, 318331 (1992).
474, 473482 (1994). 40. DiFrancesco, D. Dual allosteric modulation of pacemaker (f) channels by
25. Pape, H.-C. Queer current and pacemaker: the hyperpolarization-activated cAMP and voltage in rabbit SA node. J. Physiol. (Lond.) 515, 367376
cation current in neurons. Annu. Rev. Physiol. 58, 299327 (1996). (1999).
26. Lthi, A. & McCormick, D. A. Periodicity of thalamic synchronized oscillations: 41. Lthi, A. & McCormick D. Modulation of a pacemaker current through
the role of calcium mediated upregulation of Ih. Neuron 20, 553563 (1998). Ca2+-induced stimulation of cAMP production. Nat. Neurosci. 2, 634641
27. BoSmith, R. E., Briggs, I. & Sturgess, N. C. Inhibitory actions of ZENECA (1999).
ZD7288 on whole-cell hyperpolarization activated inward current (If) in guinea- 42. Bobker, D. H. & Williams, J. T. Serotonin augments the cationic current Ih in
pig dissociated sinoatrial node cells. Brit. J. Pharmacol. 110, 343349 (1993). central neurons. Neuron 2, 15351540 (1989).
28. Harris, N. C. & Constanti A. Mechanism of block by ZD7288 of the 43. Kiehn, O. & Harris-Warrick, R. M. 5-HT modulation of hyperpolarization-
hyperpolarization-activated inward rectifying current in guinea pig substantia activated inward current and calcium-dependent outward current in a
nigra neurons in vitro. J. Neurophysiol. 74, 23662378 (1995). crustacean motor neuron. J. Neurophysiol. 68, 496508 (1992).
29. Dixon, D. & Atwood, H. L. Crayfish motor nerve terminals response to serotonin 44. Pape, H.-C. Adenosine promotes burst activity in guinea-pig
examined by intracellular microelectrode. J. Neurobiol. 16, 409424 (1985). geniculocortical neurones through two different ionic mechanisms.
30. Dudel, J. The effect of polarizing current on action potential and transmitter J. Physiol. (Lond.) 447, 729753 (1992).
2000 Nature America Inc. http://neurosci.nature.com

release in crayfish motor nerve terminals. Pflgers Arch. 324, 227248 (1971). 45. Wojtowicz, J. M. & Atwood, H. L. Presynaptic membrane potential and
31. McCormick, D. A. & Pape, H.-C. Properties of a hyperpolarization-activated transmitter release at the crayfish neuromuscular junction. J. Neurophysiol.
cation current and its role in rhythmic oscillation in thalamic relay neurones. 52, 99113 (1984).
J. Physiol. (Lond.) 431, 319342 (1990). 46. Hubbard, J. I. & Willis, W. D. Hyperpolarization of mammalian motor nerve
32. Maccaferri, G., Mangoni, M., Lazzari, A. & DiFrancesco, D. Properties of the terminals. J. Physiol. (Lond.) 163, 115137 (1962).
hyperpolarization-activated cation current in rat hippocampal CA1 pyramidal 47. Birch, B. D., Kocsis, J. D., DiGregorio, F., Bhisitkul, R. B. & Waxman, S. G. A
cells. J. Neurophysiol. 69, 21292136 (1993). voltage and time-dependent rectification in rat dorsal spinal root axons.
33. Pape, H.-C. Specific bradycardic agents block the hyperpolarization-activated J. Neurophysiol. 66, 719728 (1991).
cation current in central neurons. Neuroscience 59, 363373 (1994). 48. Eng, D. L., Gordon, T. R., Kocsis, J. D. & Waxman, S. G. Current-clamp
34. Travagli, R. A. & Gillis, R. A. Hyperpolarization-activated currents, Ih and IKIR, analysis of a time-dependent rectification in rat optic nerve. J. Physiol.
in rat dorsal motor nucleus of the vagus neurons in vitro. J. Neurophysiol. 71, (Lond.) 421, 185202 (1990).
13081317 (1994). 49. Grafe, P., Quasthoff, S., Grosskreutz, J. & Alzheimer, C. Function of the
35. Magee, J. C. Dendritic Ih normalizes temporal summation in hippocampal hyperpolarization-activated inward rectification in nonmyelinated
CA1 neurons. Nat. Neurosci. 2, 508514 (1999). peripheral rat and human axons. J. Neurophysiol. 77, 421426 (1997).

nature neuroscience volume 3 no 2 february 2000 141


2000 Nature America Inc. http://neurosci.nature.com

articles

Synaptic activity modulates


presynaptic excitability
Teresa A. Nick1,2 and Angeles B. Ribera1

1 Department of Physiology and Biophysics, The University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
2 Present address: Division of Biology, 216-76, California Institute of Technology, Pasadena, California 91125, USA
Correspondence should be addressed to T.A.N. (teresa@etho.caltech.edu)

Synaptic activity modulates synaptic efficacy and is important in learning and development. Here we
show that development of excitability in presynaptic motor neurons required synaptic activation of
postsynaptic muscle cells. Synaptic blockade broadened action potentials and decreased repetitive
2000 Nature America Inc. http://neurosci.nature.com

firing of presynaptic neurons. Consistent with these findings, synaptic blockade also decreased
potassium-current density in the presynaptic cell. Application of neurotrophin-3, but not related
neurotrophins, prevented these changes. Recordings from patches of somatic membrane indicated
that modifications of presynaptic potassium and sodium currents occurred in a remote, nonsynaptic
compartment. Thus, activity-dependent postsynaptic signals modulated presynaptic excitability,
potentially regulating transmission at all synapses of the presynaptic cell.

Electrical activity regulates cell death, neurite outgrowth, neu- excitability by postsynaptic cells would allow targets to modulate
ronal differentiation and synaptic competition in the developing the responsiveness of their presynaptic neurons to afferent inputs.
nervous system14. Although developmental changes in postsy- Such regulation could have profound implications for the function
naptic activity during synapse formation have been examined5, of neural networks in development and learning. Aspects of this
regulation of excitability in presynaptic cells during development work have previously appeared in abstract form (T.A.N. and
and other forms of plasticity is not well understood. Moreover, A.B.R., Soc. Neurosci. Abstr. 25, 406.18, 1999).
it is completely unknown whether activation of postsynaptic cells
regulates presynaptic excitability. This lack of understanding is RESULTS
particularly striking in light of the huge body of data on synapse- We used neuromuscular junctions (NMJs) in Xenopus neu-
specific plasticity610, which cannot explain the spread of synap- ron/myocyte co-cultures19 to investigate the role of the postsy-
tic modulation that occurs in many neural systems1114. naptic cell in the differentiation of presynaptic excitability. These
Studies suggest that the intrinsic excitability of a single neuron cultures are essentially homogeneous during the initial differen-
may be regulated according to its history of firing action poten- tiation of excitability, when duration of action potentials dra-
tials1517. Real and model neurons can respond actively to exper- matically decreases20,21. However, these cultures contain a variety
imentally induced alterations in input frequency by changing of neuronal subtypes22. We hypothesized that, as neurons mature
expression of voltage-dependent ion channels. The mechanism in these co-cultures, they become heterogeneous with regard to
underlying a neurons ability to calculate how these properties electrical excitability. In addition, we proposed that synaptic acti-
should be altered and what aspect of its activity is important in vation induces myocytes to release a retrograde signal that alters
this calculation are not known. Brain-derived neurotrophic fac- motor neuron excitability. Myocytes were the ideal postsynaptic
tor (BDNF) is proposed to be involved18, although the source of cell for this investigation because they are morphologically dis-
this factor (pre-, post-, auto- or non-synaptic) and the mecha- tinguishable from other cells and contain a variety of factors that
nism underlying its regulation of excitability are unclear. Others affect motor neurons2327. To test our hypotheses, we first com-
propose that the neuron averages over time a sequence of guess- pared the excitability of neurons that formed NMJs with
es regarding how it should respond to given inputs16. Alterna- excitability of those without NMJs. We then examined neuron
tively, a neuron may respond to signals relayed from the excitability following blockade of synaptic activity with the nico-
postsynaptic cell that indicate successful synaptic transmission. tinic acetylcholine receptor blocker -bungarotoxin (-BgTx)
Here we report that one aspect of activity important in regulating during NMJ formation and differentiation.
neuronal excitability is the extent to which such activity induces
synaptic transmission and subsequent retrograde signaling from Neuronal properties correlate with synaptic contact
activated targets. We found that neurons that did not contact muscle (solitary)
Although synaptic transmission critically depends on the shape and those that contacted muscle but did not form an NMJ (no
and frequency of presynaptic action potentials, nothing is known NMJ) were significantly different from cells that formed func-
concerning the role of synaptic activity in their regulation. Here we tional NMJs (Table 1; Figs. 1 and 2). Compared with neurons
present evidence that presynaptic action potentials can be mod- lacking NMJs (solitary, no NMJ analyzed separately), cells with
ulated by synaptic activation of efferent targets. Control of net- NMJs had higher membrane capacitance (p < 0.0001), hyper-
work properties through feedback regulation of presynaptic polarized resting potential (p < 0.0002), shorter falling phases

142 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 1. Neurons with NMJs had


a b shorter-duration action potentials
than other neurons. Traces show
sample whole-cell current-clamp

falling duration (ms)


recordings of motor neurons with

Action potential
functional NMJs (a) or myocyte
contact but no NMJ (b). Action
potentials had significantly shorter
falling durations in neurons with
NMJs compared with cells lacking
80 mV NMJs. *Significantly different from
control NMJ.

Control Control
Control NMJ Control no NMJ NMJ (29) no NMJ (28)

of the action potential (p < 0.04; Fig. 1), shorter refractory peri- NMJs fundamentally differ from cells that do not contact mus-
ods (compared only with no NMJ; p < 0.005; Fig. 2) and cle and from those that contact muscle but do not form func-
increased potassium-current (IK) density (stepped from 80 to 0 tional synapses. We next investigated whether these differences
2000 Nature America Inc. http://neurosci.nature.com

mV; p < 0.04). These findings indicate that neurons that form were due to intrinsic, cell-autonomous mechanisms of motor

Table 1. Properties of neurons vary with synaptic contact and function.


Control, with NMJ Control, no NMJ Control, solitary -BgTx, NMJ NT-3 + -BgTx, NMJ
Soma diameter (m) 23.0 0.3 23.2 0.3 23.2 0.3 22.5 0.3 23.9 0.4
n = 97 n = 85 n = 85 n = 136 n = 54

Membrane capacitance (pF) 64 4 30 1* 25 1* 50 3* 47 5*


n = 126 n = 136 n = 29 n = 175 n = 50

Input resistance (M) 723 64 836 132 689 137 681 61 646 75
n = 28 n = 13 n=6 n = 36 n = 15

Resting potential (mV) 51 2 41 2* 31 2* 43 1* 45 2*


n = 96 n = 54 n = 12 n = 111 n = 36

Mean rheobase (nA)


at 40 mV 0.65 0.03 0.60 0.04 0.54 0.05 0.80 0.05* 0.68 0.04
n = 28 n = 27 n=9 n = 45 n = 13
at 80 mV 1.64 0.05 1.52 0.04 1.55 0.07 1.57 0.03 1.48 0.05*
n = 30 n = 28 n = 10 n = 54 n = 20

Action potential falling duration


at 40 mV (ms) 1.2 0.1 1.7 0.2* 1.7 0.2* 1.7 0.1* 1.2 0.1
n = 28 n = 27 n=9 n = 45 n = 13
at 80 mV (ms) 1.2 0.1 1.8 0.2* 1.8 0.3* 2.1 0.2* 1.1 0.1
n = 29 n = 28 n = 10 n = 54 n = 20

Action potential amplitude


at 40 mV (mV) 58 2 58 3 49 4* 53 2* 53 2
n = 28 n = 27 n=9 n = 45 n = 13
at 80 mV (mV) 102 1 103 2 99 2 96 1* 96 2*
n = 29 n = 28 n = 10 n = 54 n = 20

Refractory period (ms) 13.2 1.0 20.4 2.7* 17.0 2.1 17.7 1.5* 12.2 1.3
n = 27 n = 15 n=6 n = 32 n = 10

Ik density at 0 mV (step 89 13 50 9* 51 9* 59 7* 109 15


from 80 mV; A/F) n = 17 n = 17 n = 11 n = 30 n = 17
Abbreviations: NMJ, neuromuscular junction; -BgTx, -bungarotoxin; NT3, neurotrophin-3. *Significantly different from control NMJ.

nature neuroscience volume 3 no 2 february 2000 143


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 2. Neurons with NMJs showed enhanced ability to fire repetitively.


(a) Refractory period was measured with a twin-pulse protocol. Three a
superimposed episodes from a sample cell are shown for each condi-
tion. Dotted lines indicate the end of the second current pulse for each
episode. (b) Refractory period (minimum time between two pulses that
each evoke an action potential) was shorter in neurons with NMJs com-
pared with those lacking NMJs. *Significantly different from control NMJ.

40 mV

neurons or whether signals from the postsynaptic muscle cell


induce a specific set of changes in innervating neurons.

Synaptic blockade alters motor neuron excitability Control NMJ Control no NMJ
Chronic blockade of the NMJ with rhodamine-conjugated -BgTx
changed passive (Table 1) and active (Table 1; Figs. 37) electro-
physiological properties of motor neurons. Compared with
untreated controls, motor neuron resting potential was signifi- b
cantly depolarized by chronic -BgTx treatment
2000 Nature America Inc. http://neurosci.nature.com

Refractory period (ms)


(p < 0.0001). In addition, synaptic blockade with -BgTx increased
the duration (p < 0.006; Fig. 3) and decreased the amplitude (p <
0.003) of the action potential. Similar changes in resting potential
and action potentials are observed during NMJ formation and dif-
ferentiation in vivo28,29. Thus, synaptic blockade seems to prevent
normal electrophysiological development of motor neurons.
Capacitance of motor neurons was also decreased by -BgTx
treatment (p < 0.0001; Table 1). Decreased capacitance seemed
to reflect changes in the process and/or synaptic terminal, as soma
diameter was unaffected. NMJ blockade with curare modestly Control Control
NMJ (27) no NMJ (15)
reduces the number of synapses formed at the earliest stages
examined30. Because we examined the initial stages of NMJ for-
mation, our results parallel these previous findings. The increase in action potential falling duration resulting from -
BgTx-induced synaptic blockade (Fig. 3) revealed a regulatory mech-
NT-3 prevents effects of synaptic blockade anism that may explain previous reports of synaptic depression upon
If synaptic blockade prevents release of a retrograde signal and sub- caged-calcium release in the myocyte14,32. During normal NMJ devel-
sequent differentiation of presynaptic excitability, then co-application opment without -BgTx or with the putative retrograde signal NT-
of the putative signal to -BgTx-treated junctions should overcome 3 in addition to -BgTx, the spike substantially narrows, decreasing
the effects of synaptic inactivity. Because this neurotrophin alters the amount of transmitter released per action potential. Upregulation
presynaptic secretion23,25,26,31 and is upregulated by myocyte depo- of calcium in the myocyte increases NT-3 transcription31 and, pre-
larization31, NT-3 was the first candidate retrograde signal investi- sumably, release26,31. Thus, caged-calcium release in the myocyte
gated. Co-application of NT-3 blocked effects of -BgTx on resting may induce release of NT-3, which would tend to narrow the presy-
potential (versus -BgTx alone; p < 0.002) and action potential naptic action potential and seem to produce synaptic depression.
falling duration (versus -BgTx alone; p < 0.008; Table 1; Fig. 3) but Ability of motor neurons to fire repetitively increases during
not capacitance or action potential amplitude. Thus NT-3, a poten- development33,34. Spike narrowing may allow neurons to recover
tial retrograde signal released from myocytes, alters a specific sub- from action potentials faster and thus enhance repetitive firing.
set of presynaptic electrophysiological properties. Consistent with this hypothesis, we found that synaptic blockade

a b
falling duration (ms)
Action potential

80 mV

Control -BgTx NT3 +


Control -BgTx NT3 + -BgTx
(29) (54) -BgTx (20)

Fig. 3. Chronic synaptic blockade with -BgTx induced broadening of action potentials that was prevented with neurotrophin-3. (a) Whole-cell cur-
rent-clamp recordings of motor neurons with functional NMJs following chronic -BgTx revealed dramatic broadening of action potentials that was
prevented with simultaneous application of NT-3. (b) Action potential broadening induced by -BgTx was due to an increase in the falling duration.
NT-3 prevented the -BgTx-induced increase in falling duration (indistinguishable from controls). *Significantly different from controls.

144 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 4. Synaptic blockade reduced the ability to fire


repetitively, whereas co-application of NT-3 a 10 mV
1.5 nA
increased repetitive-firing capacity to control levels.
(a) Refractory period was measured with a twin- 5 ms
pulse protocol. Three episodes from a sample cell
are superimposed for each condition. The dotted
lines indicate the end of the second current pulse for
each episode. Note that action potentials overshoot
the stimulus artifact in control and NT-3 + -BgTx
conditions. (b) Refractory period was increased by 40 mV
-BgTx but unchanged by co-applied -BgTx and
NT-3. *Significantly different from controls.
Control -BgTx NT3 + -BgTx

with -BgTx, which produces spike broaden-


ing, does indeed decrease capacity for repetitive
b

Refractory period (ms)


firing as measured by an increase in refractory
period (Fig. 4, p < 0.02). NT-3, which prevents
2000 Nature America Inc. http://neurosci.nature.com

-BgTx-induced spike broadening, also pre-


vents the increase in refractory period (versus
-BgTx; p < 0.05). These changes in repetitive
firing globally regulate neuronal output. Thus, a
change initiated by postsynaptic feedback at one
synapse may modulate the efficacy of all synaps- Control -BgTx NT3 +
es of a given presynaptic neuron. (27) (32) -BgTx (10)

NT-3 effects on IK are specific


Changes in action potential duration suggest changes in IK, which To further test the hypothesis that IK was modulated by neu-
is the ionic current primarily responsible for repolarization during an rotrophins, we exposed cultures to K252a (0.2 M), which, at
action potential. We found that chronic synaptic blockade with - this concentration, is a relatively specific inhibitor for the neu-
BgTx decreased IK density in motor neurons (measured from 20 rotrophin-receptor protein-tyrosine kinases (Trks)25,35. In the
to 0 mV; p < 0.04; Fig. 5). Moreover, NT-3 prevented this decrease in absence of synaptic blockade, K252a decreased IK compared with
current density (versus -BgTx, 20 to +10 mV; p < 0.007). untreated controls (p < 0.01; Fig. 5c). This further indicates the
role of neurotrophins in the modulation of
a I K. To investigate potential non-specific
neurotrophic effects of NT-3, we attempted
to prevent -BgTx effects on IK with two
2 nA related neurotrophins, nerve growth factor
(NGF) and brain-derived neurotrophic fac-
20 ms tor (BDNF). Neither of these factors was
effective in preventing -BgTx-induced
changes in IK (NGF versus control at 0 mV,
p < 0.04; BDNF, p < 0.05; Fig. 5c). Thus,
NT-3 was the only neurotrophin tested that
was capable of reversing the effects of
synaptic blockade on IK.
Control -BgTx NT3 + -BgTx

b c
Current density (A/F)

Fig. 5. Decreases in delayed-rectifier IK density


induced by synaptic blockade were prevented by
Current density
at 0 mV (A/F)

co-application of NT-3, but not other neu-


rotrophins. (a) Sample whole-cell patch-clamp
NT3 + -BgTx (17) recordings of IK in motor neurons with NMJs
Control (17) revealed a substantial decrease in IK induced by
-BgTx (30) chronic -BgTx that was prevented by NT-3.
(b) Current-densityvoltage plots show that at
several voltages -BgTx induced a current
decrease that was prevented by NT-3.
NT3 + -BgTx (17)

K252a (10)
NGF + -BgTx (5)
BDNF + -BgTx (7)
Control (17)

-BgTx (30)

(c) Although prevented by NT-3, decreases in


current density induced by -BgTx were unaf-
fected by BDNF or NGF. K252a alone decreased
current densities to levels comparable to those
Voltage (mV)
of motor neurons treated with -BgTx alone.
*Significantly different from controls.

nature neuroscience volume 3 no 2 february 2000 145


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 6. -BgTx and NT-3 shifted the voltage dependence of activation of


IK. (a) The relative conductancevoltage curve for motor neuron IK a
shifted to more depolarized values with -BgTx-induced synaptic block-

Relative tail current


ade. This shift was prevented by NT-3. (b) Boltzmann fits of GV plots
revealed a significant change in V1/2 with -BgTx that was prevented by
co-application of NT-3.

The observed changes in IK density could be due to changes


in the number of channels expressed, the properties of channels or
both. If the number of channels in the plasma membrane changed,
the maximum conductance (and, thus, maximum current den-
sity) of the neuron should also change. We found that the number
of channels, as measured by maximum whole-cell tail-current
density, was not significantly different across groups (control, 56 Voltage (mV)
6.2 A/F, n = 16; -BgTx, 44 3.9 A/F, n = 27; NT-3 + -BgTx,
53 6.4 A/F, n = 12). We next examined changes in channel prop-
erties through comparison of normalized conductancevoltage
2000 Nature America Inc. http://neurosci.nature.com

(GV) relationships for control, -BgTX and NT-3 + -BgTx b


groups. Channel properties changed in response to -BgTx, as
indicated by a shift in the GV curve compared with controls
(measured from 20 to +10 mV; p < 0.02; Fig. 6a). Moreover, NT-
3 prevented this shift (versus -BgTx, 10 to +10 mV; p < 0.05). V1/2(mV)
To determine whether this shift was due to a change in the voltage
that gave a half-maximal conductance (V1/2) and/or a change in
the slope (k) of the GV curve, we fit data from each neuron with
the Boltzmann equation. Slope values were not significantly dif-
ferent (control, 15 1.2 mV; -BgTx, 13 0.6 mV; NT-3 + -
BgTx, 14 1.2 mV). In contrast, we found that V1/2 was shifted
to a more depolarized potential by -BgTx (p < 0.0005; Fig. 6b)
and that this shift was prevented by co-application of NT-3 (ver-
sus -BgTx; p < 0.03). The finding that -BgTx and NT-3 both
affected the same IK parameters further indicates that they act to whole-cell experiments, -BgTx shifted the GV curve in
upon the same pathway. macropatches to more depolarized potentials (measured from 0
to +30 mV; p < 0.03; Fig. 7b). In these experiments, we also
Synaptic blockade alters channels in a cell-wide manner noted an overall depolarizing shift in both groups compared to
To determine whether the effects of synaptic blockade extend whole-cell data, most likely due to a difference in C-type inac-
beyond the synapse, we examined IK properties in outside-out tivation between excised membrane patches and whole-cell
macropatches pulled from somata. We found that, comparable patches 36,37. The finding that macropatches pulled from the
soma show an effect initiated at the synapse suggests
a that this particular postsynaptic effect on presynap-
tic differentiation is cell-wide.
We next asked whether synaptic blockade might
globally affect other ionic currents. One candidate was
sodium current (I Na), as the observed decrease in
action potential amplitude with -BgTx (Table 1) sug-
gested modulation of this current. In macropatches
pulled from the soma, we found that the voltage
dependence of activation of INa was shifted to more
depolarized potentials in neurons after synaptic block-
ade with -BgTx (measured at 10 mV: p < 0.04; Fig.
8). Whole-cell data obtained with curare blockade of
b the NMJ yielded similar results (T.A.N. and A.B.R.,
unpublished observations).
g/gmax

Fig. 7. GV relation of IK shifted in response to -BgTx in


macropatches pulled from the somata of neurons as in
whole-cell recordings. (a) Sample recordings of IK in
macropatches pulled from motor neuron somata. (b) GV
plots of IK from control and -BgTx-treated motor neurons.
*Significantly different from controls.

146 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 8. The GV relation of INa in macropatches


a pulled from the neuronal soma showed an -
BgTx-induced shift. (a) Sample recordings of INa
in macropatches pulled from motor neuron
somata. (b) GV plots of INa in macropatches
from control and -BgTx-treated motor neu-
rons. *Significantly different from controls.

ciently stimulates a given myocyte would


become more excitable and, thus, more
successfully activate other contacted
myocytes. This type of regulation may be
b particularly important during target-
dependent motor neuron death, during
which neurons seem to compete for limit-
g/gmax

ed muscle-derived factors42.
Studies suggest that neurons alter their
2000 Nature America Inc. http://neurosci.nature.com

excitability in response to firing histo-


ry15,16,43. A potential mechanism for this
phenomenon is provided by our data,
which indicate that a neuron changes its
excitability based upon its recent success
at synaptic output, which depends on
action potentials. Cell-wide activity-depen-
dent modulation of presynaptic excitabil-
ity by postsynaptic targets would provide
DISCUSSION a feedback mechanism through which the postsynaptic cell can
Our data indicate that motor neuron excitability is functionally modulate not only a given presynaptic neurons responsiveness
regulated by postsynaptic target cells during development. This is to inputs, but also its output efficiency across all synapses, thus
by no means a passive feedback circuit, as postsynaptic myocytes altering output to other postsynaptic cells. This type of regula-
must be synaptically activated for modulation of excitability to tion would have important effects on neural processing. Blocking
occur. In contrast to well-described synapse-specific plasticity610, transmission at only a subset of synapses would test this hypoth-
the changes in excitability we report are global. Global regula- esis and might yield further information on potential underly-
tion of excitability that actively depends on feedback cues from ing mechanisms.
target cells represents a mechanism whereby neural systems may
self-organize and function. This mechanism would allow presy- METHODS
naptic neurons to modify all outputs simultaneously in response Animals and cell culture. Xenopus laevis embryos were produced by stan-
to signals from postsynaptic cells. dard in-vitro fertilization techniques 44 and staged according to
Nieuwkoop and Faber45. Neuron/myocyte cultures were prepared as
Interestingly, the process we describe may underlie the spread
described20. The dorsal-posterior region, which contained the pre-
of retrograde modulation of presynaptic transmission reported in sumptive spinal cord and surrounding somites, was removed from neur-
the Xenopus NMJ culture system14. However, this group investi- al tube stage (stage 1719) embryos and dissociated in divalent cation-free
gated short-term modulation, whereas we investigated long-term medium (116.7 mM NaCl, 0.67 mM KCl, 0.4 mM EDTA and 4.6 mM
effects. Thus, the studies cannot be directly compared. Further Tris at pH 7.8). Motor neurons from these embryonic stages have never
examination of the temporal aspect of neurotrophin efficacy contacted muscle46. Co-cultures were plated on plastic dishes in a com-
should reveal another level of complexity in the neu- pletely defined medium (116.7 mM NaCl, 0.67 mM KCl, 1.31 mM
rotrophinexcitability relationship. MgSO4, 10 mM CaCl2 and 4.6 mM Tris at pH 7.8) and recorded 1526 h
Although neurotrophins alter synaptic efficacy and neuronal after plating. Immediately after plating, 50 ng per ml NT-3, BDNF, NGF
excitability3840, the mechanisms through which physiological (Alomone, Jerusalem, Israel), 0.2 M K252a (Calbiochem, San Diego,
California) and rhodaminated -BgTx (2 g per ml; Molecular Probes,
control of neurotrophic effects is functionally achieved are not
Eugene, Oregon) were added. Cultures were thus exposed to these
well understood40. Our data indicate that synaptic activity criti- reagents for 1526 h before recording.
cally regulates retrograde signaling between myocyte and neu-
ron, and that the activity-dependent retrograde signal may be a Electrophysiology. Current- and voltage-clamp records were obtained
specific neurotrophin, NT-3. by whole-cell patch clamp47 with 13 M electrodes. In control cultures,
In the context of neuromuscular development, cell-wide motor neurons with NMJs were identified by their ability to cause
changes in excitability due to postsynaptic activity may provide myocyte contraction upon stimulation with 6 voltage steps (60 ms each,
the soma and all synapses of the motor neuron with a measure in 10-mV increments from +10 to +60 mV) from a holding potential of
40 mV. Neurons that did not cause contraction were placed in the no-
of its success at competing in the periphery. Myocytes subject to
NMJ group. Cells that did not contact muscle were placed in the solitary
polyneuronal innervation may differentially regulate the excitabil- group. Only neurons that clearly had no contact with other neurons were
ity of motor neurons based on synaptic efficacy, as previous stud- used. In rhodaminated--BgTx-treated cultures, neurons with NMJs
ies suggest that retrograde signaling is localized to the site of were identified by their ability to cause postsynaptic clustering of nicotinic
synaptic activation32 and that presynaptic depolarization facili- acetylcholine receptors (AChRs), observed as patches of rhodamine stain-
tates neurotrophin-induced effects41. A motor neuron that effi- ing under the presynaptic terminal46. Synaptic blockade was confirmed

nature neuroscience volume 3 no 2 february 2000 147


2000 Nature America Inc. http://neurosci.nature.com

articles

by absence of myocyte contraction under motor neuron stimulation. To The pipet solution contained 95 mM CsCl, 5 mM NaCl, 0.64 mM
verify that differences in method of identifying motor neurons in con- CaCl2, 2 mM EGTA and 10 mM HEPES at pH 7.4. Neurons were held
trol versus -BgTx-treated cultures did not affect our results, we repeat- at 80 mV and stepped from 40 to +40 mV in 9 steps (10 mV, 20 ms)
ed experiments with the AChR blocker curare, which could be washed with a 5-s recovery period between steps.
out, thus allowing us to identify motor neurons by ability to produce
myocyte contraction. Under these conditions, results were similar to
those obtained with -BgTx (T.A.N. & A.B.R., Soc. Neurosci. Abstr. 24, ACKNOWLEDGEMENTS
122.3, 1998). We thank W. J. Betz, J. W. Karpen, J. L. Lubischer, T. C. Rich, K. R. Svoboda and
Although previous studies conflict on whether AChRs on motor neu- B. G. Wallace for reading the manuscript; and B. Lu, T. J. Carew, L. K.
rons are -BgTx-sensitive48,49, both studies used neurons older than those Kaczmarek and M.-M. Poo for comments and suggestions. This work was
examined here. In contrast, neurons of the same age showed no acetyl- supported by NIH grants to T.A.N. and A.B.R.
choline response50. Because the motor neurons we examined did not express
functional AChRs, they were probably not directly affected by -BgTx.
The pipet solution for current clamp and potassium current records RECEIVED 8 SEPTEMBER; ACCEPTED 19 NOVEMBER 1999
contained 104 mM KCl, 3 mM MgCl2 and 10 mM HEPES at pH 7.4.
For current-clamp recording, the bath solution contained 125 mM
1. Schilling, K., Dickinson, M. H., Connor, J. A. & Morgan, J. I. Electrical
NaCl, 3 mM KCl, 10 mM CaCl2 and 5 mM HEPES at pH 7.4. Mem- activity in cerebellar cultures determines Purkinje cell dendritic growth
brane potential was forced to 40 and 80 mV with injected current. patterns. Neuron 7, 891902 (1991).
Action potentials were initiated by 2-ms current injections, increased 2. Spitzer, N. C. A developmental handshake: neuronal control of ionic currents
in 7 increments of 0.25 nA from 0.5 to 2.0 nA, with 5-s recovery periods and their control of neuronal differentiation. J. Neurobiol. 22, 659673 (1991).
2000 Nature America Inc. http://neurosci.nature.com

between stimuli. Only action potentials that overshot the stimulus arti- 3. Ruthazer, E. S. & Stryker, M. P. The role of activity in the development of
long-range horizontal connections in area 17 of the ferret. J. Neurosci. 16,
fact by at least 2.5 mV were included in analyses. Falling duration was 72537269 (1996).
measured as the time from the peak of the action potential to half-peak. 4. Sherrard, R. M. & Bower, A. J. Role of afferents in the development and cell
The minimum current that could reliably evoke action potentials with survival of the vertebrate nervous system. Clin. Exp. Pharmacol. Physiol. 25,
a 5-s recovery period was used in a twin-pulse examination of relative 487495 (1998).
5. Sanes, J. R. & Lichtman, J. W. Development of the vertebrate neuromuscular
refractory period at 40 mV. The interval between the two pulses was junction. Annu. Rev. Neurosci. 22, 389442 (1999).
varied from 2 to 32 ms. Relative refractory period was defined as the 6. Malinow, R. Transmission between pairs of hippocampal slice neurons:
shortest interval between pulses that allowed an action potential in quantal levels, oscillations, and LTP. Science 252, 722724 (1991).
response to the second pulse. Only depolarizations that overshot the 7. Huang, Y. Y., Colino, A., Selig, D. K. & Malenka, R. C. The influence of prior
stimulus artifact by at least 2.5 mV were considered action potentials. synaptic activity on the induction of long-term potentiation. Science 255,
730733 (1992).
Input resistance was determined by a 500-ms current pulse of 0.01 or 8. Castro-Alamancos, M. A., Donoghue, J. P. & Connors, B. W. Different forms
0.1 nA. Capacitance was determined from voltage-clamp recordings. of synaptic plasticity in somatosensory and motor areas of the neocortex. J.
The membrane potential was held at 40 mV and stepped to 90 mV in Neurosci. 15, 53245333 (1995).
6 steps of 10 mV. The area of the uncompensated capacitative current 9. Davis, G. W. & Goodman, C. S. Synapse-specific control of synaptic efficacy
(charge) was plotted against voltage and fit with a linear regression. at the terminals of a single neuron. Nature 392, 8286 (1998).
10. Martin, K. C. et al. Synapse-specific, long-term facilitation of Aplysia sensory
The slope of this line gave the capacitance. All data were analyzed with to motor synapses: a function for local protein synthesis in memory storage.
a two-tailed Students t-test and reported as mean standard error. Cell 91, 927938 (1997).
Data were judged significant if p < 0.05. Numbers of observations are 11. Vincent, P. & Marty, A. Neighboring cerebellar Purkinje cells communicate
indicated in parentheses. via retrograde inhibition of common presynaptic interneurons. Neuron 11,
885893 (1993).
For IK records, the bath solution contained 80 mM NaCl, 3 mM 12. Otani, S., Connor, J. A. & Levy, W. B. Long-term potentiation and evidence
KCl, 5 mM MgCl2, 10 mM CoCl2 and 5 mM HEPES at pH 7.4. for novel synaptic association in CA1 stratum oriens of rat hippocampus.
Tetrodotoxin (1 M; Calbiochem) was added to block voltage-gated Learn. Mem. 2, 101106 (1995).
sodium currents. Neurons were held at 80 mV and stepped from 50 13. Muller, D., Hefft, S. & Figurov, A. Heterosynaptic interactions between LTP
to +100 mV in 16 steps (10 mV, 60 ms) with a 5-s recovery period and LTD in CA1 hippocampal slices. Neuron 14, 599605 (1995).
14. Cash, S., Zucker, R. S. & Poo, M. M. Spread of synaptic depression mediated
between steps. To minimize the possibility of inadequate space clamp, by presynaptic cytoplasmic signaling. Science 272, 9981001 (1996).
cells with capacitances above 35 pF were not used for IK analyses; this 15. Desai, N. S., Rutherford, L. C. & Turrigiano, G. G. Plasticity in the intrinsic
cut-off was selected because currentcapacitance plots show a decline excitability of cortical pyramidal neurons. Nat. Neurosci. 2, 515520 (1999).
in measured current density when capacitance is 40 pF (data not 16. Stemmler, M. & Koch, C. How voltage-dependent conductances can adapt to
maximize the information encoded by neuronal firing rate. Nat. Neurosci. 2,
shown). The -BgTx-induced GV shift found in whole-cell record- 521527 (1999).
ings was also found in macropatches, suggesting that poor control of 17. Spitzer, N. C. New dimensions of neuronal plasticity. Nat. Neurosci. 2,
membrane voltage in the whole-cell recordings was not responsible 489491 (1999).
for the altered GV relationship. Records were filtered at 5 kHz and 18. Desai, N. S., Rutherford, L. C. & Turrigiano, G. G. BDNF regulates the
sampled at 25 kHz with pClamp 6.2 (Axon, Foster City, California). intrinsic excitability of cortical neurons. Learn. Mem. 6, 284291 (1999).
19. Henderson, L. P., Smith, M. A. & Spitzer, N. C. The absence of calcium blocks
For current-density measurements, mean steady-state current was impulse-evoked release of acetylcholine but not de novo formation of
measured 5060 ms after the initiation of the voltage step. Tail current functional neuromuscular synaptic contacts in culture. J. Neurosci. 4,
was measured 0.6 ms after the end of the pulse, which was beyond the 31403150 (1984).
compensated capacitative artifact for all neurons included. Relative 20. Spitzer, N. C. & Lamborghini, J. E. The development of the action potential
tail current was used as a measure of whole-cell conductance because mechanism of amphibian neurons isolated in culture. Proc. Natl. Acad. Sci.
USA 73, 16411645 (1976).
this measure obviates considerations of driving force. Calculations of 21. Henderson, L. P. & Spitzer, N. C. Autonomous early differentiation of
conductance from steady-state current yielded similar results (data neurons and muscle cells in single cell cultures. Dev. Biol. 113, 381387
not shown). For macropatch recordings, the signal-to-noise ratio was (1986).
not as high, and thus conductance was calculated from the steady- 22. Bixby, J. L. & Spitzer, N. C. The appearance and development of
neurotransmitter sensitivity in Xenopus embryonic spinal neurones in vitro. J.
state current. Maximum tail-current density was used as a measure of
Physiol. (Lond.) 353, 143155 (1984).
channel number because the headstage of our amplifier (Axopatch 23. Lohof, A. M., Ip, N. Y. & Poo, M.-M. Potentiation of developing
1C, Axon) was saturated by the large steady-state currents in control neuromuscular synapses by the neurotrophins NT-3 and BDNF. Nature 363,
NMJs, and because this measure removes driving-force considera- 350353 (1993).
tions. Inherent in the use of current density as a measure of channel 24. Stoop, R. & Poo, M.-M. Potentiation of transmitter release by ciliary
neurotrophic factor requires somatic signaling. Science 267, 695699
number is the assumption that single-channel conductance does not (1995).
change. For INa records, the bath solution contained 105 mM NaCl, 20 25. Wang, T., Xie, K. & Lu, B. Neurotrophins promote maturation of developing
mM TEA-Cl, 3 mM KCl, 10 mM CoCl2 and 5 mM HEPES at pH 7.4. neuromuscular synapses. J. Neurosci. 15, 47964805 (1995).

148 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

26. Liou, J.-C. & Fu, W.-M. Regulation of quantal secretion from developing 38. Lesser, S. S., Sherwood, N. T. & Lo, D. C. Neurotrophins differentially
motoneurons by postsynaptic activity-dependent release of NT-3. J. Neurosci. regulate voltage-gated ion channels. Mol. Cell. Neurosci. 10, 173183
17, 24592468 (1997). (1997).
27. Wang, X. H. & Poo, M. M. Potentiation of developing synapses by 39. Sherwood, N. T., Lesser, S. S. & Lo, D. C. Neurotrophin regulation of ionic
postsynaptic release of neurotrophin-4. Neuron 19, 825835 (1997). currents and cell size depends on cell context. Proc. Natl. Acad. Sci. USA 94,
28. Kellerth, J. O., Mellstrom, A. & Skoglund, S. Postnatal excitability changes of 59175922 (1997).
kitten motoneurones. Acta Physiol. Scand. 83, 3141 (1971). 40. McAllister, A. K., Katz, L. C. & Lo, D. C. Neurotrophins and synaptic
29. Ziskind-Conhaim, L. Electrical properties of motoneurons in the spinal cord plasticity. Annu. Rev. Neurosci. 22, 295318 (1999).
of rat embryos. Dev. Biol. 128, 2129 (1988). 41. Boulanger, L. & Poo, M.-M. Presynaptic depolarization facilitates
30. Dahm, L. M. & Landmesser, L. T. The regulation of synaptogenesis during neurotrophin-induced synaptic potentiation. Nat. Neurosci. 2, 346351 (1999).
normal development and following activity blockade. J. Neurosci. 11, 42. Purves, D. & Lichtman, J. W. in Principles of Neural Development 131154
238255 (1991). (Sinauer, Sunderland, Massachusetts, 1985).
31. Xie, K., Wang, T., Olafsson, P., Mizuno, K. & Lu, B. Activity-dependent 43. Golowasch, J., Abbott, L. F. & Marder, E. Activity-dependent regulation of
expression of NT-3 in muscle cells in culture: implications in the potassium currents in an identified neuron of the stomatogastric ganglion of
development of neuromuscular junctions. J. Neurosci. 17, 29472958 (1997). the crab Cancer borealis. J. Neurosci. 19, RC33 (1999).
32. Cash, S., Dan, Y., Poo, M.-M. & Zucker, R. Postsynaptic elevation of calcium 44. Tabti, N. & Poo, M.-M. in Culturing Nerve Cells (eds. Banker, G. & Goslin, K.)
induces persistent depression of developing neuromuscular synapses. Neuron 137154 (MIT Press, Cambridge, Massachusetts, 1991).
16, 745754 (1996). 45. Nieuwkoop, P. D. & Faber, J. Normal Table of Xenopus laevis (Daudin,
33. Fulton, B. P. & Walton, K. Electrophysiological properties of neonatal rat Amsterdam, 1967).
motoneurones studied in vitro. J. Physiol. (Lond.) 370, 651678 (1986). 46. Kullberg, R. W., Lentz, T. L. & Cohen, M. W. Development of the myotomal
34. Xie, H. & Ziskind-Conhaim, L. Blocking Ca2+-dependent synaptic release neuromuscular junction in Xenopus laevis: an electrophysiological and fine-
delays motoneuron differentiation in the rat spinal cord. J. Neurosci. 15, structural study. Dev. Biol. 60, 101129 (1977).
59005911 (1995). 47. Hamill, O. P., Marty, A., Neher, E., Sakmann, B. & Sigworth, F. J. Improved
35. Berg, M. M., Sternberg, D. W., Parada, L. F. & Chao, M. V. K-252a inhibits patch-clamp techniques for high-resolution current recording from cells and
2000 Nature America Inc. http://neurosci.nature.com

nerve growth factor-induced trk proto-oncogene tyrosine phosphorylation cell-free membrane patches. Pflugers Arch. 391, 85100 (1981).
and kinase activity. J. Biol. Chem. 267, 1316 (1992). 48. Perrins, R. & Roberts, A. Nicotinic and muscarinic ACh receptors in
36. Marom, S., Goldstein, S. A., Kupper, J. & Levitan, I. B. Mechanism and rhythmically active spinal neurones in the Xenopus laevis embryo. J. Physiol.
modulation of inactivation of the Kv3 potassium channel. Receptors Channels (Lond.) 478, 221228 (1994).
1, 8188 (1993). 49. Fu, W. M., Liou, H. C. & Chen, Y. H. Nerve terminal currents induced by
37. Kupper, J., Bowlby, M. R., Marom, S. & Levitan, I. B. Intracellular and autoreception of acetylcholine release. J. Neurosci. 18, 99549961 (1998).
extracellular amino acids that influence C-type inactivation and its 50. Bixby, J. L. & Spitzer, N. C. The appearance and development of
modulation in a voltage-dependent potassium channel. Pflugers Arch. 430, chemosensitivity in Rohon-Beard neurones of the Xenopus spinal cord. J.
111 (1995). Physiol. (Lond.) 330, 513536 (1982).

nature neuroscience volume 3 no 2 february 2000 149


2000 Nature America Inc. http://neurosci.nature.com

articles

A new form of long-term


depression in the perirhinal cortex
K. Cho1, N. Kemp1, J. Noel1,2, J. P. Aggleton3, M. W. Brown1 and Z. I. Bashir1

1 MRC Centre for Synaptic Plasticity, Dept. of Anatomy, University of Bristol, Bristol BS8 1TD, UK
2 Present address: Biologie cellulaire des compartiments calciques, Universite de Nice-Sophia Antipolis, Faculte des sciences, 06108 Nice, France
3 School of Psychology, Cardiff University, Cardiff CF10 3YJ, UK
Correspondence should be addressed to Z.I.B. (z.i.bashir@bris.ac.uk)

We demonstrate a form of long-term depression (LTD) in the perirhinal cortex that relies on interac-
tion between different glutamate receptors. Group II metabotropic glutamate (mGlu) receptors
2000 Nature America Inc. http://neurosci.nature.com

facilitated group I mGlu receptor-mediated increases in intracellular calcium. This facilitation plus
NMDA receptor activation may be necessary for induction of LTD at resting membrane potentials.
However, depolarization enhanced NMDA receptor function and removed the requirement of
synergy between group I and group II mGlu receptors: under these conditions, activation of only
NMDA and group I mGlu receptors was required for LTD. Such glutamate receptor interactions
potentially provide new rules for synaptic plasticity. These forms of LTD occur in the perirhinal
cortex, where long-term decreases in neuronal responsiveness may mediate recognition memory.

There are good reasons for believing that decreases in synaptic of cortex. Here we describe conditions for the induction of activ-
efficacy in neurons of the perirhinal cortex, the region of tem- ity-dependent LTD in adult perirhinal cortex. We demonstrate
poral cortex adjacent to the rhinal sulcus, are related to recogni- a new form of LTD in this cortex that relies on the activation of
tion memory1. Lesions of perirhinal cortex in rats and primates NMDA and group I and group II mGlu receptors.
impair performance of recognition-memory tasks24. Importantly,
perirhinal neuronal responses to novel visual stimuli decrease RESULTS
markedly once such stimuli become familiar1,5,6. The decrement In the perirhinal cortex, stimulation delivered to either side of the
in responsiveness can last many hours, and this change may pro- rhinal sulcus in layers II/III resulted in synaptic transmission that
vide the information required to solve such tasks. Activity-depen- was dependent on AMPA/kainate and NMDA receptors30 (Fig. 1).
dent long-term depression of synaptic transmission in perirhinal Low-frequency stimulation (LFS; 200 stimuli, 1 Hz) delivered to
cortex provides a potential mechanism for explaining decreases in either the entorhinal or the temporal side of the rhinal sulcus
neuronal responsiveness. combined with depolarization of the postsynaptic neuron to 40
Induction of homosynaptic LTD depends on postsynaptic mV induced robust homosynaptic LTD measured 3035 minutes
increases in calcium79 brought about by different mechanisms after stimulation. No differences were found between the magni-
that include activation of NMDA receptors1012 or mGlu recep- tude of LTD with stimulation to the entorhinal or the temporal
tors1317. However, there is no evidence for a role of mGlu recep- cortex side (p > 0.05); therefore, data from the two inputs were
tor activation under conditions in which NMDA receptor pooled (mean depression of 47 8%, n = 10; Fig. 2a) in this and
activation underlies the induction of activity-dependent LTD. subsequent experiments. Voltage clamp of neurons at 40 mV for
Furthermore, there seems to be no role for NMDA receptor acti- 200 s without LFS did not result in LTD (+1 8%; n = 8, p > 0.05;
vation when mGlu receptors are involved in the induction of data not shown). LTD was also induced by LFS delivered while
activity-dependent LTD. Group I mGlu receptors and NMDA the postsynaptic cell was voltage clamped at 70 mV (depression
receptors cause distinct forms of LTD by coupling to the activa- of 37 8%, n = 8; Fig. 2b). LTD induced by either of the above
tion of protein kinase C and protein phosphatases, respective- protocols was maintained for as long as the recording was con-
ly1416,18. An alternative potential means of LTD induction by tinued (up to 180 min; data not shown).
group I mGlu receptor activation is through phosphoinositide- The NMDA receptor antagonist AP5 (50 M) blocked the
induced release of calcium from intracellular stores1922. A presy- induction of LTD measured 3035 min after LFS delivered at 40
naptic role for group II mGlu receptors in LTD at mossy fiber mV (2 5%, n = 7, p > 0.05; Fig. 2c). In three experiments, AP5
synapses23,24 seems to depend on cAMP turnover25, but the mech- was washed out, and subsequent LFS resulted in LTD (52 11%,
anisms underlying the role of group II mGlu receptors in other n = 3, p < 0.05; Fig. 2c). In a separate series of experiments, AP5
regions are not well understood2628. blocked LTD at 70 mV (1 5%, n = 7; p > 0.05; Fig. 2d) in a
Perirhinal cortex is critically involved in a number of different reversible manner (48 9%, p < 0.05, n = 3; Fig. 2d). In keep-
types of memory. NMDA receptor-dependent LTP29,30 can be ing with the voltage dependence of NMDA receptor-mediated
induced in perirhinal cortex in vitro; in addition, investigating synaptic transmission, LTD was prevented by hyperpolarizing
mechanisms of LTD may provide insights into fundamental neurons to 90 mV (7 6%, n = 3, not shown) or 110 mV
mechanisms of learning and memory in this and other regions (3 11%, n = 3, not shown). To determine if a postsynaptic rise

150 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a required for the induction of LTD. This result was unexpected


because in previously reported cases of activity-dependent LTD,
induction relies on either NMDA or mGlu receptors but not on
activation of both.
The activation of group II metabotropic glutamate (mGlu)
receptors by application of DCG-IV can result in a long-lasting
depression of field EPSPs in perirhinal cortex36. We investigated
whether synaptic activation of group II mGlu receptors has a role
in LTD. The group II mGlu receptor antagonist EGLU37 did not
block the induction of LTD when LFS was delivered at 40 mV
(depression of 40 9%, n = 4; p < 0.01, Fig. 4a). Surprisingly,
however, EGLU blocked LTD when LFS was delivered at 70 mV
(depression of 3 7%, n = 7; p > 0.05; Fig. 4b). In 3 experiments,
b EGLU was washed out, and subsequent LFS induced LTD (46
12%, n = 3, p < 0.05; Fig. 4b). LTD was also blocked by the group
II/III mGlu receptor antagonist CPPG (200 M) when LFS was
delivered at 70 mV (2 10% in CPPG and 47 14% following
washout; n = 3; data not shown).
2000 Nature America Inc. http://neurosci.nature.com

Because the role of group II mGlu receptors in LTD depend-


ed on membrane potential, we investigated whether group II
mGlu receptors were involved in LTD under protocols in which
the membrane potential was not voltage clamped, thus more
closely approximating conditions in vivo. LFS was delivered while
the postsynaptic neuron was held in current clamp at 70 mV, a
c value close to normal resting potential. With the membrane
potential free to change during LFS, LTD was still reliably induced
(depression of 39 7%, n = 3; p < 0.05, Fig. 4c). Interestingly,
under these conditions LTD still depended on the activation of
group II mGlu receptors, as LTD was blocked by EGLU (3 14%,
Fig. 1. The perirhinal cortex. (a) Lateral view of the rat brain (from ref. n = 3; p > 0.05, Fig. 4d).
44) indicating the position of the perirhinal cortex (areas 35 and 36) to The voltage dependence of involvement of group II mGlu
either side of the rhinal sulcus. Approximate angles of in vitro slices are receptors in LTD was unexpected. We reasoned that group II
illustrated by diagonal lines. (b) Representation of the perirhinal cortex mGlu receptors might be important during decreased NMDA
slice indicating electrode positions. Whole-cell recordings (Rec) were receptor activation. Therefore, their involvement might relate to
obtained from single neurons in layer II/III. Stimulating electrodes (S1 voltage dependence of NMDA receptor activation, and not to a
and S2) were positioned in layer II/III either side (temporal and entorhi- voltage dependence of group II mGlu receptors per se. If this were
nal) of the recording electrode. (c) Synaptic EPSCs in perirhinal cortex
the case, increasing NMDA receptor activation and calcium influx
neurons voltage clamped at 70 mV rely on activation of AMPA/kainate
and NMDA receptors, as demonstrated by the effects of NBQX and at 70 mV should remove the requirement for group II mGlu
AP5 (ref. 30). receptors in LTD induction. In keeping with this suggestion, when
extracellular calcium was increased (from 2 to 4 mM), LTD
induced at 70 mV was not blocked by EGLU (63 7%, n = 3; p
< 0.01, Fig. 5a). When extracellular magnesium was reduced
in calcium was necessary for LTD, we used whole-cell filling solu- (from 1 to 0.01 mM) to enhance NMDA receptor function, LTD
tion containing 10 mM EGTA. Under these recording conditions, induced at 70 mV was not blocked by EGLU (41 9% depres-
induction of LTD was prevented when LFS was delivered at either sion, n = 3; p < 0.01, Fig. 5b). Thus enhancing NMDA receptor
40 or 70 mV (Fig. 2e and f). activation removes group II mGlu receptor involvement in LTD.
To determine if metabotropic glutamate (mGlu) receptors Conversely, decreasing NMDA receptor activation at 40 mV
have a role in LTD, we used the broad-spectrum (group I/II) might produce a requirement for group II mGlu receptor activa-
mGlu receptor antagonist MCPG31,32. MCPG (500 M) prevented tion in LTD at depolarized membrane potentials. In a low con-
the induction of LTD when LFS was delivered at 40 mV (Fig. centration of AP5 (2 M), LTD was induced by pairing LFS with
3a; 1 6%, n = 7, p > 0.05). Following MCPG washout in 3 depolarization of the postsynaptic neuron to 40 mV. LTD
experiments, LFS resulted in LTD (47 7%, n = 3, p < 0.05; Fig. induced at 40 mV under these conditions was blocked by EGLU
3a). MCPG also blocked induction of LTD at 70 mV (Fig. 3b; (+6 10%, measured 10 min after LFS; n = 3; p > 0.05, Fig. 5c
1 9%, n = 7, p > 0.05) in a reversible manner (46 6%, n = 3, and d). These results suggest that the levels of NMDA receptor
p < 0.05; Fig. 3b). The group I mGlu receptor antagonist activation determine the requirement of group II mGlu receptors
AIDA33,34 blocked LTD at 40 mV (Fig. 3c; +1 7%, n = 7, p > in LTD.
0.05) in a reversible manner (35 7%, n = 3, p < 0.05). At 70 One explanation for these results is that cooperativity between
mV, AIDA also produced a block of LTD induction (Fig. 3d; 1 group I and II mGlu receptors and NMDA receptors may be
11%, n = 6, p > 0.05) that was reversible (35 10%, n = 2). required to increase calcium levels for induction of LTD at rest-
MAP4, a group III mGlu receptor antagonist35, failed to prevent ing membrane potentials. Because LTD is blocked during antag-
LTD at either 40 or 70 mV (43 11%, and 41 10%, respec- onism of either NMDA or group I mGlu receptors and because
tively, n = 4, p < 0.05 for each; Fig. 3e and f). These results suggest group II mGlu receptors do not directly couple to calcium sig-
that both group I mGlu receptors and NMDA receptors are naling, it is possible that group II mGlu receptors interact with

nature neuroscience volume 3 no 2 february 2000 151


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 2. LTD relies on activation of


a b NMDA receptors and a postsynaptic
increase in calcium. (a) LFS paired
with depolarization to 40 mV results
in LTD in the input receiving LFS but
Normalized peak EPSC

not in the control input (not shown).


(% of baseline)

(b) Delivering LFS with the neuron


voltage clamped at 70 mV also results
in homosynaptic LTD, with no effect
on the control input (not shown). (c)
LTD is blocked by the NMDA recep-
tor antagonist AP5 when LFS is paired
with depolarization to 40 mV. LFS fol-
lowing AP5 washout in three of these
Time (min) Time (min) experiments induced LTD. (d) LTD
was blocked by AP5 when LFS was
c d delivered at 70 mV. In three experi-
ments, LFS induced LTD after AP5 was
washed out. (e, f) The calcium chela-
Normalized peak EPSC

tor EGTA (10 mM) blocks the induc-


2000 Nature America Inc. http://neurosci.nature.com

(% of baseline)

tion of LTD 2025 min after LFS at


both 40 mV (e) and 70 mV (f). The
control concentration of EGTA (0.5
mM) had no effect on LTD in inter-
leaved control experiments (not
shown). Illustrated synaptic responses
are taken from appropriate time
points, as indicated (1, 2). The control
and depressed responses (and voltage
Time (min)
Time (min)
steps; 5 mV) are also shown superim-
posed (1, 2). The period of LFS (200
e f stimuli, 1 Hz) is indicated by the hori-
zontal bar joined by the two arrows. In
Normalized peak EPSC

this and subsequent figures, filled cir-


cles indicate experiments in which the
(% of baseline)

neuron was depolarized to 40 mV


only for the duration of LFS. Open cir-
cles indicate experiments in which LFS
was delivered while the neuron was
voltage clamped at 70 mV. Breaks in
the x axes (c, d) indicate the time at
which AP5 was washed out.

Time (min) Time (min)

NMDA or group I mGlu receptors. We demonstrated a syner- (Fig. 6c). In separate experiments (Fig. 6c and e), the enhance-
gistic interaction between group II and group I mGlu receptors ment by DCG-IV of DHPG-induced calcium mobilization
(Fig. 6). At 70 mV, the inward current produced by the group (44 7%; n = 23 neurons) was reversibly blocked by the group II
I mGlu receptor agonist DHPG37 was significantly (p < 0.05) mGlu receptor antagonist EGLU (15 4%; n = 23 neurons).
increased by the group II mGlu receptor agonist DCG-IV (Fig. 6a; Therefore, group II and group I mGlu receptors can interact to
15 6 without DCG-IV; 59 17 pA with DCG-IV, n = 4). increase intracellular calcium levels in neurons of the perirhinal
Because DCG-IV acts as an agonist at NMDA receptors38 (albeit cortex. We postulate that this synergistic increase in calcium may
at higher concentrations than 0.5 M), these experiments were underlie the observed induction of LTD at resting membrane
carried out in the presence of 50 M AP5. In control experiments potentials.
without DCG-IV, a second application of DHPG produced
inward currents of similar magnitudes to that produced by a first DISCUSSION
application of DHPG (data not shown). This study describes an activity-dependent LTD in the adult
To investigate how interaction between group I and group II perirhinal cortex. This finding is significant because this LTD
mGlu receptors might facilitate LTD, we monitored intracellular may reflect mechanisms underlying recognition memory-related
calcium levels. In cultured perirhinal cortex neurons, DHPG (20 decreases in neuronal responses in perirhinal cortex in vivo1,5,6.
M) increased fluorescence of Fluo-3-AM filled neurons The mechanisms of homosynaptic activity-dependent LTD in
(21 6% increase; n = 16 neurons). This was significantly (p < the perirhinal cortex are notable because these depend on synap-
0.05) enhanced (45 10%; n = 16, Fig. 6b and d) by co-applica- tic activation of both NMDA receptors and mGlu receptors, in
tion of DHPG with the group II mGlu receptor agonist DCG-IV marked contrast to previously described forms of activity-depen-
(1 M). DCG-IV itself had no effect on fluorescence levels dent LTD that depend either on the activation of NMDA recep-

152 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 3. LTD depends on group I but


a b not group III mGlu receptor activa-
tion. (a) LTD is blocked by the mGlu
Normalized peak EPSC

receptor antagonist MCPG when


(% of baseline)

LFS is delivered at 40 mV. In three


experiments, MCPG was washed
out, and LTD was subsequently
induced. (b) LTD was blocked by
MCPG at 70 mV; in 3 experiments,
LFS resulted in LTD following
washout. (c) The group I mGlu
receptor antagonist AIDA prevented
the induction of LTD at 40 mV; in
Time (min) Time (min) three experiments, LFS after AIDA
washout resulted in LTD. (d) LTD
c d was blocked by AIDA at 70 mV. In
two experiments, AIDA was washed
out and LTD subsequently induced
Normalized peak EPSC

by LFS. (e, f) LTD was not blocked


by MAP4 either at 40 mV (e) or at
(% of baseline)
2000 Nature America Inc. http://neurosci.nature.com

70 mV (f).

ever, at resting membrane poten-


tials, when calcium influx
through NMDA receptor chan-
nels is restricted, the additional
Time (min) Time (min)
activation of group I mGlu
receptors is insufficient for LTD.
Under these conditions, induc-
e tion of LTD requires activation of
f group II mGlu receptors. It is not
known if this mechanism confers
Normalized peak EPSC

unique properties on perirhinal


(% of baseline)

cortical plasticity or if it also


occurs in widespread cortical
regions. Thus, although group II
mGlu receptors have a role in
LTD in other brain regions2628,
it is not known if this also occurs
at resting membrane potentials,
as reported here. Although acti-
vation of postsynaptic group II
Time (min) Time (min) mGlu receptors probably
explains our results, the possibil-
tors1012 or on the activation of mGlu receptors13,14,16,17, but not ity that presynaptic group II mGlu receptors also have some role
on the conjoint activation of both. Indeed, distinct NMDA and in LTD cannot be ruled out23,24. It is interesting to note that the
mGlu receptor-dependent forms of LTD coexist in the CA1 region mGlu receptor antagonists MCPG and EGLU reduced the depres-
of the hippocampus15. Although we have not investigated bio- sion of transmission during LFS, an effect more evident when
chemical mechanisms of perirhinal LTD, NMDA receptor-depen- LFS was delivered at 70 mV. This suggests that synaptic activa-
dent LTD relies on protein phosphatase activation18, whereas tion of mGlu receptors may transiently decrease synaptic trans-
mGlu receptor-dependent LTD depends on protein kinase C acti- mission, possibly via a presynaptic mechanism, as has been
vation14,16 in the hippocampus. The conjoint activation of NMDA demonstrated at mossy fiber synapses39. At 40 mV, the decreased
and mGlu receptors provides an additional rule for the induction driving force also results in EPSP depression, making it difficult
of synaptic plasticity. The cerebral cortex can potentially exploit to determine the effects of mGlu receptor antagonists on depres-
the variety of such rules, either to effect different types of memo- sion during LFS.
ry or to increase the conditions for memory storage. Group II mGlu receptors are located both pre- and postsy-
The most intriguing finding is that Group II mGlu receptors naptically and are known to decrease forskolin-stimulated cAMP
are involved in LTD at resting membrane potentials but not at turnover23,24. However, we provide evidence that the activation
depolarized potentials. Our results indicate that the role for group of group II mGlu receptors can increase group I mGlu receptor-
II mGlu receptors depends on the level of NMDA receptor acti- mediated calcium mobilization. This result extends previous
vation, rather than membrane potential per se. We propose that work40,41 showing that group II mGlu receptors increase group
at depolarized potentials, when NMDA receptor activation is I mGlu receptor-dependent phosphoinositide turnover. The pre-
increased, co-activation of NMDA and group I mGlu receptors cise mechanisms underlying the synergy between these groups
is sufficient and necessary to trigger the induction of LTD. How- of mGlu receptors remain to be identified. However, this inter-

nature neuroscience volume 3 no 2 february 2000 153


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 4. The role of group II


mGlu receptors in LTD is a b
voltage dependent. (a) The
group II mGlu receptor

Normalized peak EPSC


antagonist EGLU did not

(% of baseline)
block the induction of LTD
when LFS was delivered at
40 mV. (b) However, when
LFS was delivered at 70
mV, LTD was blocked by
EGLU but could be induced
in three experiments fol-
lowing washout. (c)
Delivering LFS to the neu-
ron in current clamp (at 70 Time (min) Time (min)
mV) results in LTD induc- c d
tion that is also blocked by
the group II mGlu receptor
antagonist EGLU (d).
Normalized peak EPSC

current clamp
(% of baseline)
2000 Nature America Inc. http://neurosci.nature.com

current clamp

action may occur directly


between G subunits
(activated by group II
mGlu receptors) and
Gq/11 (activated by group
I mGlu receptors)40 rather Time (min) Time (min)
than involve group II
mGlu receptor-mediated
changes in cAMP turnover41. Whatever the underlying mecha- Therefore, these results suggest a mechanism for the requirement
nism, our results demonstrate that this interaction increases intra- of synaptic activation of group II mGlu receptors in LTD.
cellular calcium. Given the essential role of calcium in the Our results demonstrate that LTD in the perirhinal cortex can
induction of synaptic plasticity, this synergy may be required for depend on several different types of glutamate receptor, thus rais-
LTD in the perirhinal cortex at resting membrane potentials. ing the question as to the physiological relevance of various mech-

a c
Normalized peak EPSC

Normalized peak EPSC


(% of baseline)

(% of baseline)

b Time (min) d Time (min)


Normalized peak EPSC

Normalized peak EPSC


(% of baseline)
(% of baseline)

Time (min) Time (min)

Fig. 5. The role of group II mGlu receptors in LTD is influenced by the level of NMDA receptor activation. (a, b) EGLU does not block LTD at 70
mV in 4 mM extracellular Ca2+ (a) or in 0.01 mM extracellular Mg2+ (b). (c, d) Single example (c) and pooled data (d) show that EGLU prevents the
induction of LTD at 40 mV when NMDA receptor activation is decreased by 2 M AP5. Induction of LTD was not blocked by this concentration of
AP5 alone (c).

154 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 6. Group II mGlu receptor activation by DCG-IV


enhances group I mGlu receptor (DHPG)-induced inward a

Change in holding
current and calcium mobilization. (a) DHPG induced an

current (pA)
inward current that was significantly enhanced by DCG-IV. To
prevent any agonist effect of DCG-IV at NMDA receptors, we
used a low concentration of DCG-IV (0.5 M) in the presence
of 50 M AP5. (b) Single example of fluorescence increase in a
cultured perirhinal cortex neuron in response to application
of 20 M DHPG. The calcium rise in response to DHPG is
synergistically increased in a reversible manner by the co-
application of DCG-IV. (c) Single example illustrates reversible Time (min)
block by EGLU of DCG-IVs enhancement of DHPG-induced
fluorescence increase. (d) Pooled data illustrate that DHPG- b c
induced calcium mobilization was significantly enhanced by
DCG-IV. (e) Pooled data show that DCG-IVs enhancement of

( % of baseline)

(% of baseline)
Fluorescence
the DHPG response is blocked by EGLU. Neither EGLU nor

Fluorescence
DCG-IV alone had any effect on fluorescence.
2000 Nature America Inc. http://neurosci.nature.com

anisms. One possibility is that the decreased neuronal


activity with stimulus repetition in perirhinal cortex1 aris- Time (s) Time (s)
es from a variety of afferent-activation patterns appro- d e
priately encoded by the various synaptic and molecular
mechanisms we identified. It is also possible that the dif-
ferent mechanisms underlying synaptic plasticity are nec-

(% of baseline)
Fluorescence
(% of baseline)
Fluorescence

essary for the variety of forms of learning and memory


involving neurons in perirhinal cortex14. In conclusion,
the results of this study identify new mechanisms of activ-
ity-dependent synaptic plasticity that rely on a function-
al interaction between different classes of synaptically
activated glutamate receptors.
METHODS
Electrophysiology. Slices of perirhinal cortex were prepared
from adult male DA rats (150270 g, 712 weeks, Bantin and
Kingman, UK). All efforts were made to minimize numbers
of animals used. Animals were anesthetized with halothane,
decapitated in accordance with the UK Animals (Scientific Procedures) expressed relative to the normalized pre-conditioning baseline. Effects
Act 1986, the brain rapidly removed and placed in ice-cold artificial of LFS were measured at appropriate time points (averaged over five
cerebrospinal fluid (aCSF; bubbled with 95% 02/5% CO2), which com- min) after delivering LFS. Data were analyzed from only one slice per rat
prised 124 mM NaCl, 3 mM KCl, 26 mM NaHCO3, 1.25 mM NaH2PO4, unless otherwise indicated. Data pooled across slices are expressed as
2 mM CaCl2, 1 mM MgSO4 and 10 mM D-glucose. A midsagittal section means s.e. and significance (p < 0.05) tested using either paired or
of the brain was made, the rostral and caudal parts were removed by sin- unpaired t-tests as appropriate. Only experiments in which there was lit-
gle scalpel cuts approximately 45 to the dorsoventral axis, and each half tle baseline drift (<10%) were included in the pooled data. Data were
was glued by its caudal end to a vibroslice stage (Campden Instruments, recorded using an Axopatch 200 amplifier (Axon Instruments, Foster
Sileby, UK). Slices (400 m) that included perirhinal, entorhinal and City, California), monitored and analyzed on line and re-analyzed off
temporal cortices were stored submerged in aCSF (2025C). A single line (W.W. Anderson, G. L. Collingridge, Soc. Neurosci. Abstr. 23, 665,
slice was placed in a submerged recording chamber (2830C; flow rate, 1997). Agonists and antagonists were applied by addition to the per-
2 ml per min) when required. Picrotoxin (5 M) was present through- fusate.
out the experiment. Blind whole-cell recordings were obtained from
neurons in layer II/III. Pipet (47 M) solutions (280 mOsm, pH 7.2) Cell culture. Perirhinal cultures were prepared according to described
comprised 130 mM cesium methyl sulfate, 8 mM NaCl, 4 mM Mg-ATP, methods42 for preparation of hippocampal cultures. Briefly, the perirhi-
0.3 mM Na-GTP, 0.5 mM EGTA, 10 mM HEPES and 6 mM QX-314. In nal cortex was removed from rats (three to five days old) and neurons
some experiments, potassium methyl sulfate was substituted for cesium recovered by enzymatic digestion and mechanical dissociation. Cells were
methyl sulfate, and QX-314 was omitted. High-EGTA filling solution plated onto coverslips in 35-mm petri dishes. Cultures were maintained
contained 10 mM EGTA, substituted for equimolar cesium methyl sul- at 37C in a 95% O2/5% CO2-humidified incubator. The culture media
fate to maintain osmolarity. One stimulating electrode was placed dor- was composed of minimal essential media (Gibco; 30 mM glucose, 2 mM
sorostrally on the temporal cortex side (area 36) and one ventrocaudally glutamine; 15 mM HEPES, 100 g per ml bovine transferrin and 30 g
on the entorhinal cortex side (area 35/entorhinal cortex) of the rhinal per ml insulin, complemented with 510% fetal calf serum). From the
sulcus. Stimuli (constant voltage) were delivered alternately to the two second day in culture, the media were supplemented with cytosine--D-
electrodes (each electrode 0.033 Hz). Neurons were voltage clamped at arabinofuranoside (2.5 M). Neurons were used for calcium-imaging
70 mV unless otherwise indicated. To induce LTD in each slice, low- studies 1430 days after plating.
frequency stimulation (LFS; 200 stimuli, 1 Hz) was delivered to only one
input. LFS was delivered at 70 mV, paired with depolarization to 40 Calcium imaging. Imaging techniques were used as described previous-
mV or delivered under current clamp at 70 mV, as appropriate for dif- ly32,43. Briefly, cells were washed three times in HBS buffer (119 mM NaCl,
ferent experiments. Alterations of membrane potential were made only 5 mM KCl, 25 mM HEPES, 33 mM glucose, 2 mM CaCl2, 2 mM MgCl2,
during the LFS. Amplitudes of the evoked EPSCs were measured and 500 nM TTX, 1 M glycine, 100 M picrotoxin, pH 7.4; osmolarity,

nature neuroscience volume 3 no 2 february 2000 155


2000 Nature America Inc. http://neurosci.nature.com

articles

300310 mOsm) and loaded with 5 M of the membrane-permeable Ca2+ depression in adult rat hippocampus. J. Physiol. (Lond.) 511, 761770 (1998).
indicator fluo-3-AM made up in 1 mg per ml bovine serum albumin/HBS 17. Fitzjohn, S. M. et al. The potent mGlu receptor antagonist LY341495
at 37C for 20 min. Cells were then washed 3 times in HBS and incubated for identifies roles for both cloned and novel mGlu receptors in hippocampal
synaptic plasticity. Neuropharmacology 37, 14451458 (1998).
30 min in a 5% CO2 atmosphere at 22C to allow for the de-esterification of 18. Mulkey, R. M., Herron, C. E. & Malenka, R. C. An essential role for protein
the flurophore. Cells were then viewed on a BioRad (Hemel Hempstead, phosphatases in hippocampal long-term depression. Science 261, 10511055
UK) MRC600 confocal microscope equipped with an argon ion laser using (1993).
standard green filter sets and perfused continuously at 2 ml per min with 19. Pin, J.-P. & Duvoisin, R. The metabotropic glutamate receptors: Structure
and functions. Neuropharmacology 34, 126 (1995).
HBS buffer to which AP5 (50 M) was added. Agonists were bath applied. 20. Conn, P. J. & Pin, J. P. Pharmacology and functions of metabotropic
Integrations of five individual images were obtained every 10 s before, dur- glutamate receptors. Annu. Rev. Pharmacol. Toxicol. 37, 205237 (1997).
ing and after agonist application. In six of seven cover slips used in this 21. Anwyl, R. Metabotropic glutamate receptors: electrophysiological properties
study, DHPG-induced fluorescence was significantly enhanced by DCG- and role in plasticity. Brain Res. Rev. 29, 83120 (1999).
IV. All neurons from these six cover slips that initially responded to DHPG 22. Reyes, M. & Stanton, P. K. Induction of hippocampal long-term depression
requires release of Ca2+ from separate presynaptic and postsynaptic
with an increase in fluorescence were used in subsequent analysis. The flu- intracellular stores. J. Neurosci. 16, 59515960 (1996).
orescence of individual cells in each preparation was measured using a pub- 23. Yokoi, M. et al. Impairment of hippocampal mossy fibre LTD in mice lacking
lic-domain NIH program (http://rsb.info.nih.gov/nihimage/) and expressed mGluR2. Science 273, 645647 (1996).
relative to baseline. The mean peak fluorescence was then calculated. 24. Domenici, M. R., Berretta, N. & Cherubini, E. Two distinct forms of long-
Drugs (all from Tocris, Bristol, UK) used were the N-methyl-D-aspar- term depression coexist at the mossy fiber-CA3 synapse in the hippocampus
during development. Proc. Natl. Acad. Sci. USA 95, 83108315 (1998).
tate (NMDA) receptor antagonist D-2-amino-5-phosphonopentanoate 25. Tzounopoulos, T., Janz, R., Sdhof, T. C., Nicoll, R. A. & Malenka, R. C. A
(AP5), the metabotropic glutamate (mGlu) receptor antagonists (S)-- role for cAMP in long-term depression at hippocampal mossy fiber synapses.
methyl-4-carboxyphenylglycine (MCPG) (R,S)-1-aminoindan-1,5-dicar- Neuron 21, 837845 (1998).
2000 Nature America Inc. http://neurosci.nature.com

boxylic acid (AIDA) (S)-2-amino-2-methyl-4-phosphonobutanoic acid 26. Huang, L. Q., Rowan, M. J. & Anwyl, R. mGluR II agonist inhibition of LTP
induction, and mGluR II antagonist inhibition of LTD induction, in the
(MAP4) and (2S)--ethylglutamic acid (EGLU) and the mGlu receptor dentate gyrus in vitro. Neuroreport 8, 687693 (1997).
agonists (R,S)-3,5-dihydroxyphenylglycine (DHPG) and (2S,2R,3R)- 27. Manahan-Vaughan, D. Group 1 and 2 metabotropic glutamate receptors play
2-(2,3-dicarboxycyclopropyl)glycine (DCG-IV). differential roles in hippocampal long-term depression and long-term
potentiation in freely moving rats. J. Neurosci. 17, 33033311 (1997).
28. Manahan-Vaughan, D. Priming of group 2 metabotropic glutamate receptors
ACKNOWLEDGEMENTS facilitates induction of long-term depression in the dentate gyrus of freely
moving rats. Neuropharmacology 37, 14591464 (1998).
We thank A. Doherty, L. Pickard and V. Collett for help with cell culture and 29. Bilkey, D. K. Long-term potentiation in the in vitro perirhinal cortex displays
calcium imaging and G.L. Collingridge for discussions. This work was supported associative properties. Brain Res. 733, 297300 (1996).
by the BBSRC, MRC and Wellcome Trust. 30. Ziakopoulos, Z., Tillet, C. W., Brown, M. W. & Bashir, Z. I. Input- and layer-
dependent synaptic plasticity in the rat perirhinal cortex in vitro.
Neuroscience 92, 459472 (1999).
RECEIVED 29 OCTOBER, ACCEPTED 23 NOVEMBER 1999 31. Eaton, S. A. et al. Competitive antagonism at metabotropic glutamate
receptors by (S)-4-carboxyphenylglycine and (R,S)--methyl-4-
carboxyphenylglycine. Eur. J. Pharmacol. 244, 195197 (1993).
1. Brown, M. W. & Xiang, J.-Z. Recognition memory: neuronal substrates of the 32. Bashir, Z. I. et al. Induction of LTP in the hippocampus needs synaptic
judgement of prior occurrence. Prog. Neurobiol. 55, 149189 (1998). activation of glutamate metabotropic receptors. Nature 363, 347350 (1993).
2. Meunier, M., Bachevalier, J., Mishkin, M. & Murray, B. Effects on visual 33. Pellicciari, R. et al. 1-aminoindan-1,5-dicarboxylic acid: a novel antagonist at
recognition of combined and separate ablations of the entorhinal and phosholipase C-linked metabotropic glutamate receptors. J. Med. Chem. 38,
perirhinal cortex in rhesus monkeys J. Neurosci. 13, 54185432 (1993). 37173719 (1995).
3. Sakai, K. & Miyashita, Y. Neural organization for the long-term memory of 34. Moroni, F. et al. Pharmacological characterization of 1-aminoindan-1,5-
paired associates. Nature 354, 152155 (1999). dicarboxylic acid, a potent mGluR1 antagonist. J. Pharmacol. Exp. Ther. 281,
4. Corodimas, K. P. & Ledoux, J. E. Disruptive effects of posttraining perirhinal 721729 (1997).
cortex lesions on conditioned fear: contributions of contextual cues Behav. 35. Salt, T. E. & Eaton, S. A. Distinct presynaptic metabotropic receptors for L-
Neurosci. 109, 613619 (1995). AP4 and CCG1 on GABAergic terminals: pharmacological evidence using
5. Zhu, X. O. & Brown, M. W. Changes in neuronal activity related to the novel -methyl derivative mGluR antagonists, MAP4 and MCCG, in the rat
repetition and relative familiarity of visual stimuli in rhinal and adjacent thalamus in vivo. Neuroscience 65, 513 (1995).
cortex of the anesthetized rat. Brain Res. 689, 101110 (1995). 36. McCaffery, B. et al. Synaptic depression induced by pharmacological
6. Zhu, X. O., McCabe, B. J., Aggleton, J. P. & Brown, M. W. Mapping visual activation of metabotropic glutamate receptors in the perirhinal cortex in
recognition memory through expression of the immediate early gene c-fos. vitro. Neuroscience 93, 977984 (1999).
Neuroreport 7, 18711875 (1996). 37. Ito, I. et al. 3,5-Dihydroxyphenylglycine: a potent agonist of metabotropic
7. Bear, M. F. & Malenka, R. C. Synaptic plasticity: LTP and LTD. Curr. Opin. glutamate receptors. Neuroreport 3, 10131016 (1992).
Neurobiol. 4, 389399 (1994). 38. Wilsch, V. W., Pidoplichko, V. I., Opitz, T., Shinozaki, H. & Reymann, K. G.
8. Kerr, D. S. & Abraham, W. C. LTD: Many means to how many ends? Metabotropic glutamate receptor agonist DCG-IV as NMDA receptor
Hippocampus 6, 3034 (1996). agonist in immature rat hippocampal neurones. Eur. J. Pharmacol. 262,
9. Bear, M. F. & Abraham, W. C. Long-term depression in hippocampus. Annu. 287291 (1994).
Rev. Neurosci. 19, 437462 (1996). 39. Scanziani, M., Salin, P. A., Vogt, K. E., Malenka, R. C. & Nicoll, R. A. Use-
10. Fujii, S., Saito, K., Miyakawa, H., Ito, K. & Kato, H. Reversal of long-term dependent increases in glutamate concentration activate presynaptic
potentiation (depotentiation) induced by tetanus stimulation of the input to metabotropic glutamate receptors. Nature 385 630634 (1997).
CA1 neurons of guinea pig hippocampal slices. Brain Res. 555, 112122 (1991). 40. Schoepp, D. D. et al. The novel metabotropic glutamate receptor agonist
11. Mulkey, R. M. & Malenka, R. C. Mechanisms underlying induction of 2R,4R- APDC potentiates stimulation of phosphoinositide hydrolysis in the
homosynaptic long-term depression in area CA1 of the hippocampus. rat hippocampus by 3,5dihydroxyphenylglycine: Evidence for a synergistic
Neuron 9, 967975 (1992). interaction between group 1 and group 2 receptors. Neuropharmacology 35,
12. Dudek, S. M. & Bear, M. F. Homosynaptic long-term depression in area CA1 16611672 (1996).
of hippocampus and effects of N-methyl-D-aspartate receptor blockade. Proc. 41. Mistry, R., Golding, N. & Challiss, R. A. J. Regulation of phosphoinositide
Natl. Acad. Sci. USA 89, 43634367 (1992). turnover in neonatal rat cerebral cortex by group I- and II-selective
13. Bashir, Z. I. & Collingridge, G. L. An investigation of depotentiation of long- metabotropic glutamate receptor agonists. Br. J. Pharmacol. 123, 581589
term potentiation in the CA1 region of the hippocampus. Exp. Brain Res. 100, (1998).
437443 (1994). 42. Noel, J. et al. Surface expression of AMPA receptors in hippocampal neurones
14. Bolshakov, V. Y. & Siegelbaum, S. A. Postsynaptic induction and presynaptic is regulated by an NSF-dependent mechanisms. Neuron 23, 365376 (1999).
expression of hippocampal long-term depression. Science 264, 11481152 43. Doherty, A. J., Collingridge, G. L. & Jane, D. E. Antagonist activity of -
(1994). substituted 4-carboxyphenylglycine analogues at group I metabotropic
15. Oliet, S. H. R., Malenka, R. C. & Nicoll, R. A. Two distinct form of long-term glutamate receptors expressed in CHO cells. Br. J. Pharmacol. 126, 205210
depression coexist in CA1 hippocampal pyramidal cells. Neuron 18, 969982 (1999).
(1997). 44. Burwell, R. D., Witter, M. P. & Amaral, D. G. Perirhinal and postrhinal
16. Otani, S. & Connor, J. A. Requirement of rapid Ca2+ entry and synaptic cortices of the rat: A review of the neuroanatomical literature and
activation of metabotropic glutamate receptors for the induction of long-term comparison with findings from the monkey brain. Hippocampus 5, 390408
(1995).

156 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Polyglutamine expansion down-


regulates specific neuronal genes
before pathologic changes in SCA1
Xi Lin1,2, Barbara Antalffy3, Dongcheul Kang1,2, Harry T. Orr4 and Huda Y. Zoghbi1,2

1 Howard Hughes Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
2 Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
3 Department of Pathology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
4 Department of Laboratory Medicine and Pathology, Institute of Human Genetics, University of Minnesota, Harvard Street at East River Rd., Minneapolis,
Minnesota 55455, USA
2000 Nature America Inc. http://neurosci.nature.com

Correspondence should be addressed to H.Y.Z. (hzoghbi@bcm.tmc.edu)

The expansion of an unstable CAG repeat causes spinocerebellar ataxia type 1 (SCA1) and several
other neurodegenerative diseases. How polyglutamine expansions render the resulting proteins
toxic to neurons, however, remains elusive. Hypothesizing that long polyglutamine tracts alter gene
expression, we found certain neuronal genes involved in signal transduction and calcium homeosta-
sis sequentially downregulated in SCA1 mice. These genes were abundant in Purkinje cells, the
primary site of SCA1 pathogenesis; moreover, their downregulation was mediated by expanded
ataxin-1 and occured before detectable pathology. Similar downregulation occurred in SCA1 human
tissues. Altered gene expression may be the earliest mediator of polyglutamine toxicity.

Spinocerebellar ataxia type 1 and several other autosomal domi- altered expression in the B05 mice3,4 using a PCR-based cDNA
nant neurodegenerative diseases are caused by expansions of a CAG subtractive-hybridization strategy. Six neuronal genes, all highly
trinucleotide repeat tract that result in an abnormally long polyg- abundant in Purkinje cells, were downregulated at a surprisingly
lutamine tract within the mutated proteins. In SCA1, the degener- early stage in pathogenesis, before any known behavioral or patho-
ation afflicts primarily the cerebellar Purkinje cells and brainstem logical changes. This downregulation required the nuclear local-
neurons, leading to the characteristic ataxic phenotype and bulbar ization of mutant ataxin-1 and occurred in two independent SCA1
dysfunction. There is persuasive evidence that the mutant proteins mouse models. We also found upregulation of one gene, a
gain some toxic function1,2, but precisely what this new function homolog of human 1-antichymotrypsin (1-ACT), at five weeks
might be and how a ubiquitous protein can cause the degeneration of age; this is most likely a secondary event in pathogenesis. Final-
of only highly circumscribed neuronal populations remain some ly, all the alterations noted in SCA1 transgenic mice were also
of the most puzzling questions in SCA1 pathogenesis. found in human SCA1 tissues. These data suggest that polygluta-
Mouse models yield important clues to this question. The B05 mine expansion in SCA1 may mediate early pathogenic events by
line, the primary SCA1 mouse model, expresses a full-length SCA1 downregulating the expression of specific neuronal genes.
cDNA with 82 CAG repeats (82Q) under the Purkinje cell-spe-
cific promoter of the Pcp2 gene and develops the ataxia and loss of RESULTS
motor coordination typical of the human disease phenotype3,4. In B05 mice, the transgene expression was first detectable at post-
These mice duplicate the Purkinje cell pathology seen in human natal day 10 (P10). The mice developed normally with no neu-
patients, including the localization of the mutant ataxin-1 pro- robehavioral abnormalities until five weeks of age, when they first
tein into a single nuclear aggregate5. A line of transgenic mice that showed mild impairment on the accelerating rotating rod. Purkinje
expresses expanded ataxin-1 with a point mutation in the nuclear cells begin to lose their proximal dendrites at six weeks3,4. To cap-
localization signal (so that the protein cannot be transported into ture the full spectrum of genes that might be altered in SCA1
the nucleus) fails to develop disease6. Another mouse line (77) pathogenesis, we performed PCR-based cDNA subtraction using
expressing expanded ataxin-1 without a self-association domain cerebellar mRNA from two-month-old B05 mice and wild-type
fails to develop nuclear aggregates despite nuclear localization of littermates. The subtractions were done in two different directions,
the expanded proteinbut does develop the SCA1 phenotype. using B05 mRNA either as the tester or the driver, to identify genes
These data clearly demonstrate that nuclear events, though not that were up- or downregulated, respectively. Six hundred clones
nuclear aggregation, mediate SCA1 pathogenesis. Furthermore, from the subtracted cDNA library were screened by dot-blot
studies in transfected COS-1 cells show that mutant ataxin-1 asso- hybridization. Twenty clones that differentially hybridized to the
ciates with nuclear matrix and disrupts specific nuclear domains subtracted probes were further examined with northern blots,
(such as promyelocytic oncogenic domains, PODs)5. from which nine clones representing seven different genes had
Disruption of nuclear structures and functions might alter gene altered expression patterns in B05 cerebellum. Differentially
expression. In testing this hypothesis, we found several genes with expressed clones were identified by sequence analysis.

nature neuroscience volume 3 no 2 february 2000 157


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 1. PCCMT expression was specifically


reduced in SCA1 mice. (a) Northern blots of a
cerebellar RNA from four different lines of
SCA1 transgenic mice and wild-type (WT) lit-
termates using PCCMT cDNA as a probe. B05
and A02 mice express human ataxin-1 with 82
and 30 glutamines, respectively. K772T mice
express expanded ataxin-1 with 82 glutamines
and a point mutation in the nuclear localization
signal (K772T). 77 mice express ataxin-1 with
77 glutamines and no self-association domain.
Substantially reduced PCCMT mRNA expres-
sion was seen in B05 and 77 mice, but not in
A02 and K772T mice. The ages of animals used
in this experiment were eight weeks for B05
and A02 and five weeks for 77 and K772T. b
The -actin hybridization signal demonstrates
equal loading of RNA for littermate pairs at
various time points. (b) Northern blots of
cerebellar RNA from B05 mice and WT litter-
2000 Nature America Inc. http://neurosci.nature.com

mates at different ages using PCCMT cDNA as


a probe. Expression of PCCMT mRNA was
reduced by postnatal day 11. The similar
amount of RNA between B05 and WT litter-
mates was shown by the equal -actin
hybridization signal.

To determine whether these altered expression levels might reduction in PCCMT mRNA level was already detectable in B05
mediate early pathogenesis, we examined the expression of these cerebella at P11 (Fig. 1b), about one day after the initiation of SCA1
genes in younger mice, before any histologic or neurobehavioral transgene expression and two weeks before the earliest morpho-
abnormalities develop. Because mutant animals do not begin to logic changes3,4. Moreover, this reduction was consistently observed
display Purkinje cell pathology until six weeks4, we reasoned that in multiple experiments using RNA from different litters of mice.
changes of gene expression before three weeks would be less likely to Phosphoimaging analysis confirmed that the reduction of PCCMT
represent secondary events in pathogenesis. We ascertained the ear- mRNA expression was statistically significant, with an 11 2.36%
liest date of altered expression for each gene by northern blot analy- decrease at day 11 and a 19.2 7.5% decrease at day 15 (n = 3, p <
ses on total-RNA samples collected at different time points from 0.05). Thus PCCMT downregulation occurred very early.
B05 mice or normal littermates. The characterization of these genes We examined PCCMT mRNA expression in other SCA1 trans-
and their alterations in SCA1 is detailed in the following sections. genic mouse lines that show no obvious Purkinje cell dysfunction
or degeneration (A02 mice and ataxin-1[82Q]K772T mice)4,6. A02
Cloning and characterization of mouse PCCMT mice express high levels of wild-type human ataxin-1 with 30 glu-
One of the clones identified by cDNA subtractive hybridization was tamines (30Q); mice expressing ataxin-1[82Q]K772T with a point
isolated repeatedly and found to contain a 550-bp insert. Sequenc- mutation in the nuclear-localization signal retain mutant ataxin-
ing and BLAST searches revealed no significant homology to any 1 in the cytoplasm, where it is nonpathogenic. Neither line showed
known sequences in the databases. Northern blots using the 550- altered PCCMT mRNA levels (Fig. 1a, A02 and K772T). In con-
bp insert as a probe identified a 4.8-kb transcript, which was dra- trast, PCCMT mRNA was dramatically decreased in another SCA1
matically downregulated in B05 cerebellum (Fig. 1a, B05). mouse model (77) that manifests behavioral and pathological
Subsequent screening of a mouse brain cDNA library (Stratagene, changes similar to those of B05 mice (Fig. 1a, 77).
La Jolla, California) allowed the isolation of a clone that contained To study the function of PCCMT in mice, we examined its
a 4,730-bp insert (GenBank accession number, AF209926). Assem- expression during development and in various peripheral tissues in
bly of this 4,730-bp sequence with that of an EST clone (AA022288) northern blots. PCCMT mRNA was highly enriched in adult cere-
resulted in a cDNA of 4,846 bp and predicted an open reading frame bellum, with low levels of expression in other brain regions (Fig.
of 852 bp that was highly homologous to the coding regions of 2a) and very low but detectable (after prolonged exposure) levels in
human (88% identity) and Xenopus (71% identity) prenylcysteine peripheral tissues such as lung, heart, kidney and skeletal muscles
carboxylmethyltransferase (PCCMT)7,8. This clone was therefore (Fig. 2b). Within the cerebellum, PCCMT mRNA was expressed
identified as a mouse homolog of PCCMT. Immunohistochem- postnatally, with a dramatic increase after P12 (Fig. 2c). The tem-
istry using a rabbit polyclonal antibody against a peptide (amino poral and spatial expression pattern of PCCMT suggests that it
acids 185200) from human PCCMT protein, equivalent to the might have an important role in Purkinje cells.
predicted mouse protein amino acids 184199, showed abundant To determine whether PCCMT was expressed in neurons vul-
staining in the Purkinje cells of wild-type mice and sparse staining nerable to SCA1, we examined PCCMT mRNA expression in the
in B05 Purkinje cells (data not shown). human brain (Fig. 2d). Northern blots using mRNA isolated from
To evaluate the potential roles of PCCMT in early SCA1 patho- different brain regions showed that PCCMT was expressed in the
genesis, we examined PCCMT mRNA expression at different time cerebellum and putamen at higher levels than in other brain
points in B05 and wild-type littermate cerebella. Remarkably, the regions and revealed three different transcripts of 9.0 kb, 5.5 kb

158 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 2. PCCMT mRNA


expression in mouse and a d
human brain. (ac) Northern
blots using PCCMT cDNA as
a probe and total RNA (25
g) from different brain
regions of adult WT mice (a),
thalamus and several periph-
eral tissues of B05 and WT
littermates at two months of
age (b), and cerebella of WT
mice at different developmen-
tal stages (c). Ethidium bro-
mide-stained RNA gel images b e
are shown for loading con-
trol, which was confirmed by
hybridization with -actin
cDNA (not shown). PCCMT
was predominantly expressed
in cerebellum, with much
2000 Nature America Inc. http://neurosci.nature.com

lower levels of expression in


other regions of the brain (a,
one-day exposure). In the
peripheral tissues, PCCMT
was detectable only after long
exposure (b, three-day expo-
sure). During cerebellar c
development, PCCMT
expression was low before P6
and dramatically increased
after P12. (d) Northern blot
of polyA RNA (2 g) from
the indicated regions of
human brain probed with
PCCMT cDNA shows the
relatively high levels of
expression in the cerebellum
and putamen. Equal loading in
each lane was confirmed by hybridization with -actin cDNA as a probe (not shown). (e) Immunohistochemical staining of PCCMT in the cerebella and pons
of a SCA1 patient and an age-matched control demonstrates much-reduced expression in Purkinje cells and pontine neurons in the patient.

and 3.6 kb, of which the 5.5 kb was most abundant (Fig. 2d). It mRNAs of IP3R1 and SERCA2 are downregulated at two weeks of
remains unknown whether these transcripts represent alterna- age in B05 mouse cerebellum, again, preceding known behavioral
tively spliced isoforms or distinct homologs. Immunohisto- or morphological changes (Fig. 3a). Immunohistochemistry using
chemical analysis using an antibody against human PCCMT either an IP3R1- or SERCA2-specific polyclonal antibody on cere-
found abundant expression in Purkinje cells and pontine neurons bellar sections from six-week-old mice revealed dramatic reductions
(Fig. 2e, control). We also examined PCCMT expression in human in IP3R1 and SERCA2 proteins in B05 cerebellar Purkinje cells (data
brain tissue from a juvenile-onset SCA1 patient, focusing on the not shown). Similar downregulations in IP3R1 and SERCA2 mRNA
cerebellum and pons, two regions typically affected in SCA1. Lit- occurred in mice expressing ataxin-1[77Q], but not in A02 mice or
tle PCCMT immunostaining was found in the cerebellum (Fig. mice expressing ataxin-1[82Q]K772T (data not shown). Therefore,
2e, Cerebellum, SCA1). This might be due in part to the paucity downregulation of IP3R1 and SERCA2 specifically occurs in the
of remaining Purkinje cells in this patient. The pons, however, transgenic mice that show Purkinje cell degeneration caused by
which retained many pontine neurons, showed greatly reduced mutant ataxin-1. Immunohistochemistry using IP3R1 (not shown)
PCCMT immunostaining (Fig. 2e, Pons, SCA1). Although it is and SERCA2 (Fig. 3b) antibodies revealed profoundly reduced
impossible to investigate the role of PCCMT in early SCA1 patho- immunostaining in cerebellar Purkinje cells and pontine neurons
genesis in humans, these snapshots of end-stage disease are in from SCA1 patient tissues.
accordance with the alterations observed in transgenic mice.
Downregulation of 5-phosphatase, mTRP3 and EAAT4
IP3R1 and SERCA2 are downregulated at P14 Three genes were downregulated by three to four weeks of age in
Sequence analysis of two other subtracted clones showed 100% iden- B05 mouse cerebellum: type 1 inositol polyphosphate 5-phosphatase
tity to mouse inositol triphosphate receptor type 1 (IP3R1) and sar- (5-phosphatase; Fig. 4a), transient receptor potential type 3 (TRP3;
coplasmic endoplasmic reticulum calcium ATPase type 2 (SERCA2), Fig. 4b) and excitatory amino acid transporter type 4 (EAAT4; Fig.
two Purkinje cell-abundant proteins. IP3R1 and SERCA2, an intra- 4c). Type 1 inositol polyphosphate 5-phosphatase is a major inac-
cellular Ca2+-release channel and a Ca2+ pump, respectively, are both tivating enzyme of IP3 and IP4, which mobilize Ca2+ through inter-
present on the membrane of the endoplasmic reticulum (ER). The actions with the intracellular calcium release channels, IP3Rs9. TRP3

nature neuroscience volume 3 no 2 february 2000 159


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 3. IP3R1 and SERCA2 expression


is reduced in SCA1 mice and patient a b
brain tissues. (a) Northern blots of
total cerebellar RNA (25 g) from
B05 mice and WT littermates at dif-
ferent ages using IP3R1 or SERCA2
cDNA as probe. IP3R1 and SERCA2
mRNAs are reduced in B05 cerebel-
lum at two weeks of age. Equal loading
of RNA in each lane is shown by
similar intensity of ethidium bro
mide-stained 28S and 18S rRNAs.
(Hybridization signal of -actin is not
shown.) (b) Immunohistochemical
staining of SERCA2 in the cerebellum
and pons of a SCA1 patient and an
age-matched control.
2000 Nature America Inc. http://neurosci.nature.com

is a putative plasmalemmal calcium channel controlled by the con- 1, as downregulation occurred in ataxin-1[77Q] mice but not
tent or filling state of calcium stores through some unknown mech- A02 or ataxin-1[82Q]K772T mice (data not shown).
anism10. Transcripts of these two genes were dramatically reduced
at four weeks of age in B05 mouse cerebellum (Fig. 4a and b). These Purkinje cell gene expression was not globally altered
downregulations probably reflected reduced expression in Purkinje To rule out the possibility that downregulation might be a global
cells, as type 1 inositol 5-phosphatase and TRP3 are predominantly effect, we looked at the expression patterns of eight other genes
expressed in Purkinje cells in the cerebellum. known to be abundant in Purkinje cells in three-month-old B05
EAAT4 is a Purkinje cell-specific glutamate transporter local- mice and wild-type littermates. Among those examined were
ized on dendritic spines11. In B05 cerebellum, EAAT4 mRNA down- cGMP binding-phosphodiesterase (cGB-PDE), phospholipase C4
regulation was obvious at three weeks of age. By six weeks, EAAT4 (PLC4) and IP3 3-kinase (IP3-3K)1214. There were no apparent
protein was hardly detectable in Purkinje cells by immunohisto- changes in the expression of any of the eight genes in B05 mouse
chemistry using an EAAT4-specific polyclonal antibody (Fig. 4c). cerebellum (three examples shown in Fig. 5).
Analysis of the expression patterns of these three genes in the
other SCA1 transgenic mice demonstrated that their downregu- Mouse 1-antichymotrypsin upregulated at five weeks
lation was associated specifically with pathogenic mutant ataxin- Not all the genes identified with our subtractive hybridization
approach were downregulated in B05 mice. One clone
was upregulated. Sequence analysis revealed that this
a clone was identical to EB22/4, a serine protease
inhibitor isolated from a mouse teratocarcinoma cell
line and presumably the physiological homolog of
human 1-antichymotrypsin (61% amino acid iden-
tity)15. Northern blots showed a single transcript of
2 kb (Fig. 6a). In B05 cerebellum, EB22/4 mRNA
b was upregulated by five weeks (Fig. 6b), whereas no
change was detected in A02 mice (Fig. 6a). EB22/4
mRNA was also increased in ataxin-1[77Q] mouse
cerebellum (Fig. 6a), consistent with the pathology
of Purkinje cell degeneration. Surprisingly, we
observed an increase in EB22/4 in ataxin-1[82Q]K772T
mice (Fig. 6a), which have no obvious Purkinje cell
c
Fig. 4. Downregulation of type 1 inositol polyphosphate 5-
phosphatase, TRP3 and EAAT4 in SCA1 mice. Northern
blots of cerebellar RNA (25 g) from B05 mice and WT lit-
termates at different ages were probed with 5-phosphatase 1
(a), TRP3 (b) and EAAT4 (c) cDNAs. Equal amount of total
RNA in each lane was confirmed by similar -actin hybridiza-
tion signals and ethidium bromide staining of the gel (not
shown). (a, b) At four weeks of age, inositol polyphosphate
5-phosphatase type 1 and TRP3 showed reduced expression
in B05 cerebellum. (c) At three weeks, EAAT4 mRNA is
reduced. Immunohistochemical staining of EAAT4 in B05 and
WT littermate mouse cerebella at six weeks of age showed
greatly reduced EAAT4 protein in Purkinje cell dendrites.

160 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

inhibitor and an acute response protein involved in a variety of


pathological conditions. In the brain, 1-antichymotrypsin is
expressed in glia; its expression is increased in Alzheimer disease,
Huntington disease and normal aging1820, and it may well be
involved in similar molecular events in these different pathophys-
iological conditions. The observation that EB22/4 was also upreg-
ulated in K772T mice indicates its dependence on the expression,
but not the nuclear localization, of mutant ataxin-1. Although
K772T mice do not exhibit abnormalities, their Purkinje cells
might respond in subtle ways to the presence of expanded ataxin-
1 in the cytoplasm. Excessive quantities of misfolded ataxin-1 in
the cytoplasm could elicit enough of a stress response to affect cel-
lular functions, if not cause actual degeneration.
Fig. 5. Mutant ataxin-1 does not affect expression of all Purkinje cell-
specific genes. Northern blots of total cerebellar RNA (25 g) from
EAAT4, TRP3 and inositol polyphosphate 5-phosphatase type
three-month-old B05 mice and WT littermates using the indicated 1 were downregulated in B05 cerebellum by three to four weeks of
cDNAs as probes. The mRNA levels of cGMP binding-phosphodi- age. EAAT4 is a Purkinje cell-specific glutamate transporter local-
esterase (cGB-PDE), phospholipase C4 (PLC4) and IP3 3-kinase (IP3- ized to the extra-junctional dendritic membrane. It presumably
3K) are similar between B05 and WT. restricts glutamate spillover to adjacent synapses and enhances
2000 Nature America Inc. http://neurosci.nature.com

spatial and temporal resolution of glutamate signaling11,21. TRP3,


a calcium channel abundant in Purkinje cell membranes, is
involved in cellular calcium homeostasis10,22,23. Inositol polyphos-
degeneration but do express mutant ataxin-1 in the cytoplasm. phate 5-phosphatase type 1 is the major inactivating enzyme of
Unlike the other genes characterized above, EB22/4 mRNA upreg- calcium-mobilizing molecules IP3 and IP4 and is involved in the
ulation thus seemed to depend on the polyglutamine expansion in regulation of calcium signaling9,24,25. In both human and rat
ataxin-1, but not on nuclear localization of its product. brains, inositol 5-phosphatase type 1 is highly abundant in the
Because EB22/4 is highly homologous to human 1-ACT and Purkinje cells12,26. Specific downregulation of inositol 5-phos-
anti-human 1-ACT polyclonal antibodies recognize a single band phatase type 1 and TRP3 suggests that calcium homeostasis may
on western blots of mouse tissues16, we examined 1-ACT-like be perturbed in SCA1 pathogenesis.
protein (EB22/4) expression by immunohistochemistry. The white At two weeks, IP3R1 expression and SERCA2 expression were
matter of B05 mouse cerebellum showed intense 1-ACT-anti- reduced in B05 mice. Both of these proteins localize to the endo-
body staining, whereas wild-type mouse cerebellum showed a very plasmic reticulum and are involved in calcium store regulation2729.
low level of staining (Fig. 6c). This staining pattern suggests that IP3R1 is a calcium-release channel and increases free cytoplasmic
upregulation of 1-ACT-like protein in SCA1 mouse cerebellum calcium concentration ([Ca2+]cyt) by releasing Ca2+ from the ER
was a response secondary to mutant ataxin-1 expression in the in response to IP3 generated by extracellular signals. Interestingly,
Purkinje cells. It is interesting that, in SCA1 patient tissue, 1- lack of IP3R1 due to either spontaneous mutation or targeted dele-
ACT stains intensely in neuronal cytoplasm (Fig. 6d). tion leads to severe ataxia, seizures and premature death in
mice30,31. SERCA2, an ATPase, balances IP3R1 by pumping calci-
DISCUSSION um into the ER, effectively reducing [Ca2+]cyt. The abundance of
We identified and characterized several specific genes whose IP3R1 and SERCA2 in cerebellar Purkinje cells reflects their impor-
expression was altered in SCA1 transgenic mice at unexpectedly tance in regulating calcium levels3234. The net effect of IP3R1 and
early stages in pathogenesis. This downregulation is the earliest SERCA2 downregulation on [Ca2+]cyt in Purkinje cells may not be
known effect of mutant ataxin-1, occurring before any detectable straightforward, however. Depending on the relative contributions
pathological changes. This effect of mutant ataxin-1 is selective: of IP3R1 and SERCA2, average [Ca2+]cyt could increase, decrease or
a number of other Purkinje cell-specific genes were not altered. not change. Even subtle changes in calcium transients might lead
Moreover, the downregulation seemed to occur sequentially, with to Purkinje cell dysfunction and progressive degeneration in
PCCMT affected first, followed by IP3R1, SERCA2, EAAT4, TRP3 SCA135,36.
and 5-phosphatase 1. The finding that downregulation of the same The cellular calcium signaling system is markedly flexible and
genes occurred in two independent SCA1 mouse models (B05 adaptable, and expression of different components are interde-
and [77Q]) and not in mice overexpressing non-pathogenic atax- pendent37. It thus seems probable that downregulating IP3R1
in-1 (A02 and mice expressing ataxin-1[82Q]K772T) makes it plau- and/or SERCA2 leads to changes in the expression of the other
sible that reduced expression of these genes is involved in SCA1 genes by a cellular compensatory mechanism that then resets the
pathogenesis. Substantially reduced immunostaining of PCCMT, cellular calcium-signaling system. Because the expression of
SERCA2 and IP3R1 in human SCA1 brain tissues corroborated expanded ataxin-1 in SCA1 mouse Purkinje cells is not transient (it
the mouse data and suggested that expression of these genes is is driven by the potent Pcp2/L7 promoter), the constant, high level
altered in human SCA1 pathogenesis as well. To rule out the of mutant ataxin-1 might pose a sustained stress signal to the cal-
remote possibility that this downregulation was secondary to cium signaling system.
Purkinje cell distress, we evaluated the expression pattern of PCCMT, whose expression was altered first, is an abundant
PCCMT, SERCA2 and TRP3 in two mouse mutants that have dys- Purkinje cell protein in both mouse and human cerebella; it is
functional Purkinje cells, Ube3a and Sca1 knockouts2,17. No alter- involved in the posttranslational modification of a number of very
ations in gene expression were detected (data not shown). important cellular proteins, including Ras-like small GTPases, het-
EB22/4 was the only gene we found to be upregulated. Avail- erotrimeric G protein subunits and nuclear lamins3840. Inhibit-
able evidence suggests that mouse EB22/4 is the physiological ing PCCMT activity impairs membrane targeting of Ras in
homolog of human 1-antichymotrypsin15, a serine protease transfected cells41. Downregulation of PCCMT in cerebellar Purk-

nature neuroscience volume 3 no 2 february 2000 161


2000 Nature America Inc. http://neurosci.nature.com

articles

a inje cells could strain the normal functions of these molecules and
have adverse effects on many aspects of cellular physiology. Alter-
natively, the reduced expression of PCCMT (as well as IP3R1 and
SERCA2) in early developmental stages might perturb Purkinje
cell differentiation and/or render the cells more susceptible to stress.
It is striking that all the genes whose expression was altered
early in the pathogenesis in SCA1 mouse models were downreg-
ulated; perhaps a common mechanism operated at transcription-
al and/or posttranscriptional levels. Because expanded ataxin-1
has to localize to Purkinje cell nuclei to cause neuronal degenera-
b tion6, we favor a model of transcriptional downregulation. Expand-
ed ataxin-1 might interact directly with certain transcription factors
and coactivators, thus interfering specifically with the transcrip-
tion of their target genes. Alternatively, expanded ataxin-1 might
affect transcription indirectly, possibly by perturbing nuclear pro-
teasomal activity42. Some transcription factors, such as NFB, p53,
nuclear receptors and their cofactors, are regulated by proteaso-
mal degradation4346. Ataxin-1 colocalizes with proteasomal com-
2000 Nature America Inc. http://neurosci.nature.com

ponents in nuclear aggregates in the Purkinje cells of both SCA1


transgenic mice and patients42. Proteasomal perturbation by
mutant ataxin-1 in the nucleus could lead to downregulation of
certain genes through either increased degradation of their acti-
vators and/or decreased degradation of their repressors, depending
c on how the proteasomal activity is modulated by expanded ataxin-
1. Regardless, identification of genes downregulated in SCA1 makes
it possible for future studies to evaluate this hypothesis.
Mutant ataxin-1 might also downregulate gene expression by
disrupting nuclear structures and nuclear protein functions. For
example, mutant ataxin-1 redistributes PML5, which interacts
with CBP/p300 and facilitates CBP/p300 localization to PODs47.
PML enhances the activity of nuclear receptors as transactivators
in mammalian cells, suggesting that it could be a transcriptional
co-activator. It is possible that, by redistributing PML, mutant
ataxin-1 interferes with its normal function as a transcriptional
co-activator, resulting in reduced expression of specific groups of
genes. It will be worthwhile to determine if the genes whose
d expression is altered by mutant ataxin-1 are regulated by nuclear
receptors and PML. Alternatively, mutant ataxin-1 might alter
gene expression by interfering with mRNA metabolism through
various nuclear events, such as mRNA processing and mRNA
nucleocytoplasmic transport. These possibilities can be explored
in the SCA1 mouse models by studying the regulation of the genes
identified in this study.
Identifying genes whose expression is altered by mutant ataxin-
1 provides a platform for further studies aimed at understanding
the earliest molecular events disrupted in SCA1 pathogenesis.
Characterization of the transcriptional regulation of these genes
should yield greater insight into not only the molecular mecha-
nisms by which expanded ataxin-1 affects gene expression, but the
pathogenesis of other polyglutamine diseases as well.

METHODS
Northern blots. Total RNA was isolated from different mouse tissues
Fig. 6. Upregulation of EB22/4, a mouse homolog of human 1-ACT. (a, with TRIZOL reagent following manufacturers instructions (Gibco-BRL,
b) Northern blots of total cerebellar RNA (25 g) from different lines of Gaithersburg, Maryland). RNA (25 g) was fractionated and transferred
SCA1 transgenic mice and WT littermates at five to eight weeks of age (a) to nylon membrane according to standard protocol. Various probes were
and B05 mice and WT littermates at different ages (b) were probed with labeled using Megaprime kit (Amersham, Cleveland, Ohio) and
EB22/4 cDNA. EB22/4 expression is increased not only in B05 and 77 hybridized in ExpressHyb hybridization solution (Clontech, Palo Alto,
mice, but also in K772T mice. The increase of EB22/4 expression in B05 California) according to manufacturers instruction. The relative amounts
cerebellum was obvious by five weeks of age. (c, d) Immunohistochemical of RNA on the blots were evaluated by both ethidium bromide-stained
staining for 1-ACT in B05 and WT littermate mouse cerebella at three RNA gel images and hybridization using -actin cDNA as a probe.
months of age (c) as well as the pons of a SCA1 patient and age-matched
control (d). Staining for 1-ACT is increased in the white matter of B05 Subtractive cDNA cloning. To identify genes with altered expression pat-
cerebellum (c) and in pontine neurons of the SCA1 patient (d). terns in B05 mice, we performed subtractive cDNA cloning using Clontech

162 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

PCR-Select cDNA Subtraction Kit (Clontech). Briefly, total RNA was iso- 16. Akaaboune, M., Ma, J., Festoff, B. W., Greenberg, B. D. & Hantai, D. Neurotrophic
lated from the cerebella of two-month-old B05 mice and wild-type, sex- regulation of mouse muscle beta-amyloid protein precursor and alpha 1-
matched littermates using TRIZOL as described above. mRNA was then antichymotrypsin as revealed by axotomy. J. Neurobiol. 25, 503514 (1994).
17. Jiang, Y. H. et al. Mutation of the Angelman ubiquitin ligase in mice causes
selected using Dynabeads Oligo(dT)25 (Dynal A. S., Oslo, Norway). We increased cytoplasmic p53 and deficits of contextual learning and long-term
then precisely followed the protocol of PCR-Select cDNA Subtraction Kit potentiation. Neuron 21, 799811 (1998).
provided by the manufacturer. 18. Abraham, C. R., Selkoe, D. J. & Potter, H. Immunochemical identification of the
Potential, differentially expressed clones were verified by northern blots serine protease inhibitor alpha 1-antichymotrypsin in the brain amyloid deposits
of Alzheimers disease. Cell 52, 487501 (1988).
of wild-type and B05 cerebellar RNA using the cDNA insert as a probe. 19. Das, S. & Potter, H. Expression of the Alzheimer amyloid-promoting factor
Sequences of the differentially expressed clones were obtained by ABI antichymotrypsin is induced in human astrocytes by IL-1. Neuron 14, 447456
PrismTM DNA Sequencer and BigDyeTM Terminator Cycle Sequencing (1995).
Ready Reaction Kit (PE Applied Biosystems, Foster City, California). The 20. Koo, E. H., Abraham, C. R., Potter, H., Cork, L. C. & Price, D. L. Developmental
sequences were analyzed by DNASTAR program and BLAST search. expression of alpha 1-antichymotrypsin in brain may be related to astrogliosis.
Neurobiol. Aging 12, 495501 (1991).
21. Tanaka, J., Ichikawa, R., Watanabe, M., Tanaka, K. & Inoue, Y. Extra-junctional
Immunohistochemistry. The immunohistochemical staining on mouse localization of glutamate transporter EAAT4 at excitatory Purkinje cell synapses.
brain sections was performed as described previously5. The rabbit poly- Neuroreport 8, 24612464 (1997).
clonal anti-PCCMT antibody, a gift from Mark Philips7, was used at 1:800 22. Birnbaumer, L. et al. On the molecular basis and regulation of cellular
capacitative calcium entry: roles for Trp proteins. Proc. Natl. Acad. Sci. USA 93,
dilution. The rabbit anti-IP3R1 antibody (sc6093, Santa Cruz, Santa
1519515202 (1996).
Cruz, California) was used at 1:100 dilution. The anti-SERCA2 mono- 23. Zhu, X. et al. trp, a novel mammalian gene family essential for agonist-activated
clonal antibody (Novocastra Laboratories, Newcastle, UK) was used at capacitative Ca2+ entry. Cell 85, 661671 (1996).
1:50 dilution. The rabbit anti-1-ACT antibody (Zymed Laboratories, 24. Drayer, A. L. et al. The family of inositol and phosphatidylinositol polyphosphate
2000 Nature America Inc. http://neurosci.nature.com

San Francisco, California) was used at 1:50 dilution. The rabbit anti- 5-phosphatases. Biochem. Soc. Trans. 24, 10011005 (1996).
25. Woscholski, R. & Parker, P. J. Inositol lipid 5-phosphatasestraffic signals and
EAAT4 antibody, provided by Kohichi Tanaka and Kei Watase11,21, was signal traffic. Trends Biochem. Sci. 22, 427431 (1997).
used at 0.1 mg per ml. 26. De Smedt, F., Verjans, B., Mailleux, P. & Erneux, C. Cloning and expression of
human brain type I inositol 1,4,5-trisphosphate 5-phosphatase. High levels of
mRNA in cerebellar Purkinje cells. FEBS Lett. 347, 6972 (1994).
ACKNOWLEDGEMENTS 27. Takei, K. et al. Ca2+ stores in Purkinje neurons: endoplasmic reticulum
subcompartments demonstrated by the heterogeneous distribution of the InsP3
The authors thank A. Beaudet for providing the Ube3a null mice, M. Philips at
receptor, Ca2+-ATPase, and calsequestrin. J. Neurosci. 12, 489505 (1992).
New York University School of Medicine and K. Tanaka at the National Institute 28. Villa, A. et al. The endoplasmic reticulum of Purkinje neuron body and
of Neuroscience in Japan for providing the anti-PCCMT and anti-EAAT4 dendrites: molecular identity and specializations for Ca2+ transport.
Neuroscience 49, 467477 (1992).
antibodies, H. J. Bellen for reading the manuscript and V. Brandt for comments.
29. Volpe, P., Nori, A., Martini, A., Sacchetto, R. & Villa, A. Multiple/heterogeneous
This work was supported by grants NS27699 and NS22920 from the National Ca2+ stores in cerebellum Purkinje neurons. Comp. Biochem. Physiol. Comp.
Institutes of Health (to H.Y.Z. and H.T.O.) and by the core facilities of the Baylor Physiol. 105, 205211 (1993).
30. Matsumoto, M. et al. Ataxia and epileptic seizures in mice lacking type 1 inositol
College of Medicine Mental Retardation Research Center. X. Lin is an Associate 1,4,5-trisphosphate receptor. Nature 379, 168171 (1996).
and H. Zoghbi an Investigator with the Howard Hughes Medical Institute. 31. Street, V. A. et al. The type 1 inositol 1,4,5-trisphosphate receptor gene is altered
in the opisthotonos mouse. J. Neurosci. 17, 635645 (1997).
32. Miller, K. K., Verma, A., Snyder, S. H. & Ross, C. A. Localization of an
RECEIVED 20 SEPTEMBER; ACCEPTED 6 DECEMBER 1999 endoplasmic reticulum calcium ATPase mRNA in rat brain by in situ
hybridization. Neuroscience 43, 19 (1991).
33. Nakanishi, S., Maeda, N. & Mikoshiba, K. Immunohistochemical localization of
1. Zoghbi, H. Y. & Orr, H. T. Spinocerebellar ataxia type 1. Seminars in Cell Biology: an inositol 1,4,5-trisphosphate receptor, P400, in neural tissue: studies in
Unstable Repeat Diseases Vol. 6, 2935 (Saunders, Philadelphia, 1995). developing and adult mouse brain. J. Neurosci. 11, 20752086 (1991).
2. Matilla, A. et al. Mice lacking ataxin-1 display learning deficits and decreased 34. Ross, C. A., Danoff, S. K., Schell, M. J., Snyder, S. H. & Ullrich, A. Three
hippocampal paired-pulse facilitation. J. Neurosci. 18, 55085516 (1998). additional inositol 1,4,5-trisphosphate receptors: molecular cloning and
3. Burright, E. N. et al. SCA1 transgenic mice: a model for neurodegeneration differential localization in brain and peripheral tissues. Proc. Natl. Acad. Sci. USA
caused by an expanded CAG trinucleotide repeat. Cell 82, 937948 (1995). 89, 42654269 (1992).
4. Clark, H. B. et al. Purkinje cell expression of a mutant allele of SCA1 in transgenic 35. Berridge, M. J. Neuronal calcium signaling. Neuron 21, 1326 (1998).
mice leads to disparate effects on motor behaviors, followed by a progressive 36. Verkhratsky, A. & Toescu, E. C. Calcium and neuronal aging. Trends Neurosci .21,
cerebellar dysfunction and histological alterations. J. Neurosci. 17, 73857395 27 (1998).
(1997). 37. Liu, B. F., Xu, X., Fridman, R., Muallem, S. & Kuo, T. H. Consequences of
5. Skinner, P. J. et al. Ataxin-1 with extra glutamines induces alterations in nuclear functional expression of the plasma membrane Ca2+ pump isoform 1a. J. Biol.
matrix-associated structures. Nature 389, 971974 (1997). Chem. 271, 55365544 (1996).
6. Klement, I. A. et al. Ataxin-1 nuclear localization and aggregation: Role in 38. Rando, R. R. Chemical biology of protein isoprenylation/methylation. Biochim.
polyglutamine-induced disease in SCA1 transgenic mice. Cell 95, 4153 (1998). Biophys. Acta 1300, 516 (1996).
7. Dai, Q. et al. Mammalian prenylcysteine carboxyl methyltransferase is in the 39. Clarke, S. Protein isoprenylation and methylation at carboxyl-terminal cysteine
endoplasmic reticulum. J. Biol. Chem. 273, 1503015034 (1998). residues. Annu. Rev. Biochem. 61, 355386 (1992).
8. Imai, Y., Davey, J., Kawagishi-Kobayashi, M. & Yamamoto, M. Genes encoding 40. Hrycyna, C. A. & Clarke, S. Modification of eukaryotic signaling proteins by C-
farnesyl cysteine carboxyl methyltransferase in Schizosaccharomyces pombe and terminal methylation reactions. Pharmacol. Ther. 59, 281300 (1993).
Xenopus laevis. Mol. Cell. Biol. 17, 15431551 (1997). 41. Choy, E. et al. Endomembrane trafficking of ras: the CAAX motif targets proteins
9. Mitchell, C. A., Brown, S., Campbell, J. K., Munday, A. D. & Speed, C. J. to the ER and Golgi. Cell 98, 6980 (1999).
Regulation of second messengers by the inositol polyphosphate 5-phosphatases. 42. Cummings, C. J. et al. Chaperone suppression of aggregation and altered
Biochem. Soc. Trans. 24, 9941000 (1996). subcellular proteasome localization imply protein misfolding in SCA1. Nat.
10. Mori, Y. et al. Differential distribution of TRP Ca2+ channel isoforms in mouse Genet. 19, 148154 (1998).
brain. Neuroreport 9, 507515 (1998). 43. Ciechanover, A. The ubiquitin-proteasome proteolytic pathway. Cell 79, 1321
11. Yamada, K. et al. EAAT4 is a post-synaptic glutamate transporter at Purkinje cell (1994).
synapses. Neuroreport 7, 20132017 (1996). 44. Masuyama, H. & MacDonald, P. N. Proteasome-mediated degradation of the
12. Mailleux, P., Takazawa, K., Erneux, C. & Vanderhaeghen, J. J. Comparison of vitamin D receptor (VDR) and a putative role for SUG1 interaction with the AF-
neuronal inositol 1,4,5-trisphosphate 3-kinase and receptor mRNA distributions 2 domain of VDR. J. Cell. Biochem. 71, 429440 (1998).
in the adult rat brain using in situ hybridization histochemistry. Neuroscience 49, 45. Nawaz, Z., Lonard, D. M., Dennis, A. P., Smith, C. L. & OMalley, B. W.
577590 (1992). Proteasome-dependent degradation of the human estrogen receptor. Proc. Natl.
13. Kotera, J. et al. Expression of rat cGMP-binding cGMP-specific Acad. Sci. USA 96, 18581862 (1999).
phosphodiesterase mRNA in Purkinje cell layers during postnatal neuronal 46. Zhang, J., Guenther, M. G., Carthew, R. W. & Lazar, M. A. Proteasomal
development. Eur. J. Biochem. 249, 434442 (1997). regulation of nuclear receptor corepressor-mediated repression. Genes Dev. 12,
14. Roustan, P. et al. The rat phospholipase C beta 4 gene is expressed at high 17751780 (1998).
abundance in cerebellar Purkinje cells. Neuroreport 6, 18371841 (1995). 47. Doucas, V., Tini, M., Egan, D. A. & Evans, R. M. Modulation of CREB binding
15. Inglis, J. D., Lee, M., Davidson, D. R. & Hill, R. E. Isolation of two cDNAs protein function by the promyelocytic (PML) oncoprotein suggests a role for
encoding novel alpha 1-antichymotrypsin-like proteins in a murine nuclear bodies in hormone signaling. Proc. Natl. Acad . Sci. USA 96, 26272632
chondrocytic cell line. Gene 106, 213220 (1991). (1999).

nature neuroscience volume 3 no 2 february 2000 163


2000 Nature America Inc. http://neurosci.nature.com

articles

High-resolution mapping of iso-


orientation columns by fMRI
Dae-Shik Kim, Timothy Q. Duong and Seong-Gi Kim

Center for Magnetic Resonance Research, University of Minnesota Medical School, 2021 6th Street S.E., Minneapolis, Minnesota 55455, USA
Correspondence should be addressed to S.-G.K.(kim@cmrr.umn.edu)

Blood-oxygenation level-dependent (BOLD) functional magnetic resonance imaging (fMRI) is an


important tool for localizing brain functions in vivo. However, the ability of BOLD fMRI to map corti-
cal columnar structures is highly controversial, as the ultimate functional specificity of BOLD remains
unknown. Here we report a biphasic BOLD response to visual stimulation in the primary visual cortex
of cats. In functional imaging, the initial BOLD signal decrease accurately labeled individual iso-
2000 Nature America Inc. http://neurosci.nature.com

orientation columns. In contrast, the delayed positive BOLD changes indicated the pattern of overall
activation in the visual cortex, but were less suited to discriminate active from inactive columns.

Localization is a principle that is widely used in brain: cytoar- resolutions), it becomes imperative to determine the limits of
chitectonically distinct areas form the basis for functional spe- the functional specificity that can be achieved with BOLD. The
cialization1. Such parcellation of the cortical tissue into functional exact temporal kinetics of the BOLD responses in mammalian
subunits is especially prominent at the level of individual cortical brains, however, remain vigorously debated (compare refs. 2426
columns. In visual cortex of mammals, neurons with similar with refs. 27, 28); thus it remains to be seen whether the func-
response properties such as ocular dominance or orientation tional specificity of BOLD is sufficient to map the basic compu-
preference are clustered into columns, spanning the entire cor- tational units of the brains functional architecture, namely, that
tical plate from the pia to white matter24. Studies of the struc- of cortical columns.
ture, function and plasticity of cortical columns using a variety
of traditional mapping techniques, however, suffer from funda- RESULTS
mental limitations. For example, intra- and extracellular record- To resolve the question of whether and to what extent the colum-
ings yield insufficient field of view, and the 2-deoxyglucose5 nar architecture of the brain can be labeled using the BOLD-
technique is not viable for mapping in vivo. The optical imaging fMRI technique, we used ultra-high field magnets to obtain MR
of intrinsic signals allows simultaneous recording of neuronal signals originating from individual orientation columns in cat
activity over large areas of cortex69. However, this technique can- visual cortex (area 18). Visual stimuli were optimized to drive
not be considered to be noninvasive, and furthermore, its appli- orientation-selective, complex-type area 18 neurons29. In this
cation is limited to the exposed cortical surface9,10. study, area 18 was used because the distance between two iso-
The progress of blood-oxygenation level-dependent func- orientation columns is greater in this area than in area 17 (ref.
tional magnetic resonance imaging11,12 raises hope that the func- 30). Furthermore, area 18 on the lateral gyrus is essentially flat
tional architecture of the living brain can be visualized in the cat and can be covered by a single imaging slice. We carried
noninvasively, avoiding the limitations of the aforementioned out a total of ten semi-chronic experiments in ten hemispheres of
techniques. Using the paramagnetic deoxyhemoglobin as an five different animals. Unless otherwise mentioned, similar results
endogenous contrast agent11,12, BOLD-based functional images were obtained in all ten experiments. Statistical data for all ten
can be obtained in vivo (in contrast to the 2-deoxyglucose (2- studies are given in parentheses.
DG) technique5,13), do not require extrinsic contrast agents (in Figure 1a shows an anatomical MR image of cat visual cor-
contrast to the positron emission technique14,15) and can access tex on the lateral gyrus. All activation maps were derived from a
activation signals from the entire brain (in contrast to optical plane tangential to area 18 on the lateral gyrus (green box,
imaging69). Most importantly, the noninvasiveness of MRI ide- Fig. 1a). Colored pixels indicating regions of increased BOLD-
ally suits this technique for studying the human brain during signal change (Fig. 1c; see Methods) reveal the pattern of cortical
cognitive and perceptual tasks1621. activation in response to a moving grating oriented at 45. As
Numerous BOLD studies during cognitive16, motor17 and per- indicated in this panel, robust and homogenous activities were
ceptual1821 tasks indicate good spatial correlation between neu- observed in the lateral gyri of both hemispheres. The region of
ronal and hemodynamic responses at a coarse scale (several activity extended several millimeters in anteriorposterior and
millimeter to centimeter), and the BOLD signal pointspread is mediallateral directions.
comparable to that of optical imaging21. The ability of BOLD In cat area 18, the average spacing between two neighboring
fMRI to map the columnar architecture of the brain, however, is iso-orientation columns is 1.21.4 mm (ref. 30). Therefore, the
controversial, as the ultimate functional specificity of BOLD is nominal in-plane resolution of 156 156 m2 per pixel achieved
undetermined. Because optical spectroscopy data predicts a in this study (see Methods) should have been sufficient to resolve
biphasic BOLD response following neuronal stimulation22,23 individual orientation columns. As evident in Fig. 1c, however, a
(with each BOLD phase potentially yielding different mapping columnar layout was not obtained. All four activation maps

164 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a b expected to greatly enhance the func-


tional specificity of fMRI. Such an
improvement of functional speci-
ficity was achieved in our study (Fig.
1d). Here, only the negative BOLD
percent changes originating within
the first two seconds after the stim-
ulus onset were used to generate
functional maps (see Methods). The
colored pixels indicate regions of sig-
nificant decrease in the MR signals
following visual stimulation. In
c d e sharp contrast to the conventional
fMRI maps based on the positive
BOLD-signal changes (Fig. 1c), the
foci of the negative signal changes are
confined to patchy clusters (Fig. 1d).
In line with the iso-orientation
2000 Nature America Inc. http://neurosci.nature.com

columns in cat detected with 2-DG30


and optical imaging32,33 techniques,
such semi-ellipsoidal and irregular-
ly shaped clusters were broadly dis-
tributed over the approximated area
18 with an average periodicity of
Fig. 1. Improvement of spatial specificity of BOLD using the initial negative signal changes. about 1.34 0.23 mm. Note also that
(a) Anatomical image of cat area 18 on the lateral gyrus (image size, 2 2 cm2). The green box indicates the areas defined by negative signal
the position of the imaging slice used to obtain the activation maps displayed in this study. (b) The bipha-
changes were located largely in tis-
sic MR signal time course following visual stimulation. Stimulus duration of 10 s is marked by the gray
box behind the time course. Scale bar (b), 10 s. (c) Pattern of increased BOLD activity in response to
sue areas, excluding regions of large
moving 45 gratings. The positive BOLD percent changes are marked with colors as indicated by the blood vessels such as that surround-
color scale below. (d) Functional map for which only negative BOLD percent changes occurring within ing the sagittal sinus.
the first two seconds after stimulus onset were used. The color scale below indicates the negative per- Given the large difference in
cent changes. (e) The pattern of positive BOLD responses after the threshold for functional image con- absolute amplitude between the neg-
struction was raised to match the number of the activated pixels to that in panel (d). See text for details. ative and positive signal changes, it
Scale bar (ce), 1 mm. A, anterior; P, posterior. is conceivable that the differences in
spatial pattern between the positive
(Fig. 1c) and negative (Fig. 1d) maps
could be simply due to the difficulty
obtained in response to moving gratings of four different orienta- in equating signal-threshold levels. Therefore, as a control, we
tions (0, 45, 90, 135) yielded homogeneous spatial distributions raised the threshold for positive responses to match the num-
that were hardly distinguishable (data not shown; see also ref. 31). ber of activated pixels to that in the negative map (Fig. 1e). The
This result is consistent with optical spectroscopy data22,23, resulting positive map largely reflects the pattern of surface vas-
suggesting that the stimulation of cortical neurons gives rise to culature in visual cortex.
a biphasic hemodynamic response of which only the early Patterns of negative activity obtained from the same patch of
increase of local deoxyhemoglobin (and hence a decrease in MR cortex while the animal viewed four different orientation stimuli
signal) reflects increased neuronal activity and oxygen con- further corroborate functional significance of the negative BOLD
sumption22,23. Subsequent decrease of deoxyhemoglobin (and signals (Fig. 2ad). Analogous to 2-DG30 and optical imaging32,33,
hence an increase of MR signal), which forms the basis for con- we show regions of high activity (large negative signals) as dark
ventional BOLD11,12, is hypothesized to originate mainly from pixels. Each pattern of negative activity was highly specific to the
the widespread increase in blood flow, thus making it less suit- respective stimulus orientation. To support this notion, outlines
able to discriminate electrically active from inactive columns. of the orientation patches for 90 and 135 maps were overlaid
In cat visual cortex, the time evolution of MR signals was indeed on the maps obtained for the respective orthogonal orientations
biphasic (Fig. 1b). Following visual stimulation (as indicated by the (Fig. 2ad).
gray box behind the time course), the MR signal decreased, reach- It is clear from these panels that the patches for orthogonal
ing a minimum of about 0.2% to 0.4% (0.28 0.1%, n = 10) orientations occupied cortical territories that were mostly com-
around 3 seconds (2.9 0.7 s, n = 10) after the stimulus onset. The plementary to each other. The complementarity between maps
signal then reversed, yielding a maximum positive signal change of of orthogonal orientations was highly significant (linear correla-
1.02.0% (1.3 0.4%, n = 10) approximately 810 seconds after tion between 0 and 90 maps, r = 0.4, p < 106; between 45 and
stimulus onset (8.0 1.3 s, n = 10). Biphasic BOLD responses fol- 135, r = 0.55, p < 106; see Methods). Stimulus-selective respons-
lowing visual stimulation were observed in all ten experiments at es based on negative BOLD signals were reflected at the colum-
two different magnetic fields. nar scale (Fig. 2e and f); these panels present the signal time
As the early negative signal changes are likely to reflect the courses obtained from 45 and 135 orientation columns (in green
transient increase of local deoxyhemoglobin in parenchymal tis- and in red, respectively; see Methods). MR signals in pixels rep-
sue 22,23, their use in generation of a functional map can be resenting 45 columns (green trace) decreased transiently during

nature neuroscience volume 3 no 2 february 2000 165


2000 Nature America Inc. http://neurosci.nature.com

articles

a b to the patchy, interdigitized columns of the negative maps, the


domains of the positive BOLD responses were larger, with no
apparent periodicity. The outlines of the 135 map from Fig.
3b were then overlaid on the 45 map (Fig. 3a). The regions of
positive activity for the orthogonal orientations were mostly
overlapping (linear correlation between the two maps, r = 0.69,
Negative BOLD activity

p < 106). The results in Figs. 2 and 3 indicate, therefore, that


although the amplitudes of the negative signals were far small-
er than those of the positive signals, the stimulus-specific con-
c d trast of the negative signals was superior to that of the positive
signals. Based on Fig. 2e and f, the stimulus-specific contrast
of the negative signals was calculated to be about 6.5 times larg-
er than that of the positive signals.
The ultimate validation of the veracity of the iso-orientation
columns based on negative BOLD changes would require simul-
taneous single-unit recording. Such simultaneous recording,
however, would require placement of the recording electrodes
e f in the iso-center of the magnetic bore, resulting in devastating
2000 Nature America Inc. http://neurosci.nature.com

magnetic field interferences. Furthermore, it would be almost


impossible to change the electrode track or introduce new elec-
Signal change (%)

trodes without repositioning the animal. Alternatively, the speci-


ficity of the negative BOLD signals at the columnar scale can be
tested by comparing their detailed spatial pattern with that
obtained by using more traditional brain-mapping techniques.
A characteristic and invariant feature of the mammalian orien-
tation system is the presence of topological singularities3234,
which are observed in many species using both multi-elec-
trode34,35 and optical-imaging32,33,3638 techniques. Therefore, to
validate the functional specificity of the negative BOLD maps,
Fig. 2. Representation of orthogonal orientations in complementary we calculated the composite-angle maps through vector sum-
cortical domains. (ad) Patterns of negative activity in response to four mation of the underlying activation maps obtained for the four
orientations (0, 45, 90, 135). Regions of negative signal changes dur-
different orientations (see Methods; Fig. 4).
ing the first two seconds following visual stimulation are displayed as
dark pixels. For visual inspection of the complementarity between the In the composite map based on only the early negative
orthogonal orientation maps, the patches in panels (b) and (d) are out- BOLD signals (Fig. 4a), the colors (preferred orientations)
lined (in blue and red, respectively) and overlaid on the maps in panels change smoothly, forming a map of orientation selectivity. The
(a) and (c), respectively. With few exceptions (marked by yellow arrow- continuity of orientation preferences is interrupted only at the
heads), the maps for orthogonal orientations are complementary to singular points (termed orientation pinwheels) where the
each other. Scale bar, 1 mm. (e, f) The signal time courses for 45 domains for all orientations converge, as observed using multi-
(green) and 135 (red) columns during the stimulation with 45 (c) and electrode34,35 and optical imaging32,33,3638 techniques. Each ori-
135 (d) orientation stimuli. The stimulation duration of ten seconds is entation is represented only once around such a pinwheel;
marked by the gray boxes behind the time courses. Scale bar (e, f), 10 s.
therefore each of these topological singularities takes on one of
two rotational chiralities, namely clockwise or counter-clock-
wise pinwheels. Two such pinwheels are enlarged (Fig. 4a, right).
The pinwheel density and the ratio between clockwise and
45 stimulation (Fig. 2e), whereas little or no such decrease was counter-clockwise singularities found in negative BOLD com-
observed during 135 stimulation (Fig. 2f). Likewise, MR signals posite maps were in excellent agreement with those obtained
from 135 columns (red trace) decreased transiently in response to with optical imaging. We found a pinwheel density of
the 135 stimulus (Fig. 2f), but not to the complementary 45 1.46 0.17 pinwheels per mm2 (n = 4); in comparison, optical
stimulus (Fig. 2e). The difference between 45 and 135 columns imaging studies 32,37 yielded average pinwheel densities of
in magnitude of the early negative signals during the 45 (Fig. 2e) 1.21.95 pinwheels per mm2. Likewise, the ratio of clockwise
and 135 (Fig. 2f) stimulation was statistically significant (p < to counterclockwise pinwheels was found to be roughly 1:1 in
0.005 and p < 0.003, respectively; paired t-test). both negative-BOLD (1:0.89, n = 4) and optical imaging 37
The positive BOLD signals (Fig. 2e and f), on the other (1:0.9) data.
hand, were much less suited to discriminate between 45 and Although the above data are in excellent agreement with data
135 columns, as they yielded largely overlapping time cours- from multi-electrode34,35 and optical imaging studies32,33,3638,
es for the two orthogonal stimuli. The differences in positive it is theoretically possible but unlikely that such topological sin-
signals originating from the orthogonal columns were statis- gularities could arise from a randomly distributed pattern of
tically insignificant (p = 0.19, paired t-test). To allow more activity39. Therefore, as a control, composite maps based on sig-
direct comparison of the map quality, we also calculated the nals obtained before stimulus onset (Fig. 4b) and those during
45 and 135 maps obtained during the positive BOLD period the delayed positive BOLD changes (Fig. 4c) were also calcu-
(Fig. 3a and b). All functional maps depicted in Figs. 2 and 3 lated. All steps for composite-map construction were performed
were acquired in the same fMRI studies, and images were exactly as for Fig. 4a (see Methods). Unlike the composite map
processed with analogous methods (see Methods). In contrast based on negative BOLD signals (Fig. 4a), the maps based on

166 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 3. Representation of orthogonal orienta-


tions of positive BOLD maps. (a, b) Functional a b
maps based on positive signal changes obtained
with 45 and 135 stimuli, respectively. High
positive signals are represented by dark pixels
(see text). Maps in Figs. 2 and 3 were obtained
from the same experiment. The domains of
high activity in 135 map (b) are outlined and
overlaid on the 45 map (a). Most of the active
domains in both maps overlapped extensively,
except a few areas as marked with green
arrowheads. Scale bar, 1 mm.

Positive BOLD activity


2000 Nature America Inc. http://neurosci.nature.com

control (Fig. 4b) or delayed positive BOLD signals (Fig. 4c) dis- the use of a differential method for BOLD fMRI data poses
played none of the characteristic features of the mammalian severe difficulties. In differential imaging, one activation map
angle maps. (for example, of the left eye) is subtracted from the other acti-
vation map (the right eye), with the assumption that the two
DISCUSSION maps yield complementary activation patterns. If this assump-
Our results indicate that the functional specificity of BOLD at tion is truesuch as between left- and right-eye domainsthen
the columnar scale depends highly on the temporal dynamics the result of the subtraction is tautological, because it was known
of the underlying signals. If the early negative signals are used, in advance. However, if the assumption is not true or simply
functional maps at columnar resolution can be obtained without unknown, as for most receptive-field properties, then the sub-
differential imaging (see below). The delayed positive BOLD traction method will probably give the wrong answer. Without
changes, on the other hand, clearly indicated the pattern of the direct validation of BOLD using simultaneous single-unit
overall activation per se in the visual cortex, but were less suit- recordings, the applicability of the differential method for BOLD
ed to discriminate between active and inactive columns, as they data remains questionable. The main significance of the nega-
were more diffuse2123 and less specific to the individual stimu- tive BOLD imaging is therefore that a columnar pattern of high
lus properties. functional specificity can be obtained directly, without the need
It is, of course, conceivable that, in principle, even such dif- for such differential methods.
fuse positive BOLD signals might yield columnar patterns of As the T2 and T2*-weighted BOLD contrast predominantly
activity if maps of orthogonal conditions were subtracted from originates from the regional changes in paramagnetic deoxyhe-
each other. Such differential imaging had been suggested for moglobin concentration10,11,42, the early decrease of BOLD signals
analyzing optical imaging40 and conventional (positive) BOLD can most likely be attributed to the regional increase of deoxyhe-
data41. Although the use of a differential method for optical- moglobin following neuronal stimulation. Although alternative
imaging data might be defendable given the well established ver- explanations exist43,44, the most parsimonious explanation for such
ification of intrinsic signals with extensive single-unit studies37,38, transient BOLD signal decrease is caused by elevated oxygen con-
sumption44 in the active orientation column without a
a commensurate increase in blood flow22,23.
Recently, similar decreases in BOLD signals have been
observed in the visual cortices of awake human2426,45 and
anesthetized nonhuman primates46 during perceptual

Fig. 4. fMRI-based composite angle maps. (a) The composite


angle map obtained through pixel-by-pixel vector addition of
the four single iso-orientation maps based on negative signal
changes. The color key next to (a) was used for color coding
the resulting orientation preferences. Overall continuity of the
b c orientation preferences is interrupted at the orientation pin-
wheels where the cortical columns for different orientations
are circularly arranged. The white and black circles in (a) depict
clockwise and counterclockwise pinwheels, respectively. Two
of such pinwheels are enlarged right (a). Scale bar for the
enlarged pinwheels, 200 m. As a control, the composite maps
based on MR signals obtained before stimulus onset (b) and
during positive BOLD signals (c) are also displayed. Maps in
(b) and (c) were obtained from the same cortical region as in
(a). The control maps are devoid of topological structures
characteristic of genuine composite angle maps. Scale bar
(ac), 1 mm. A, anterior; P, posterior; M, medial; L, lateral.

nature neuroscience volume 3 no 2 february 2000 167


2000 Nature America Inc. http://neurosci.nature.com

articles

tasks, indicating that the capability of BOLD-fMRI to label func- iso-orientation maps for each experiment. For the positive response (Fig.
tional columns in vivo should also be applicable in humans and 1c), the threshold was raised in proportion to ratio of the maximum pos-
monkeys. We conclude from our study that noninvasive fMRI of itive to the minimum negative BOLD response for each animal (approx-
brain functions can be performed at the columnar levels. Thus imately fivefold). For the negative and positive BOLD maps depicted in
it now becomes possible to study the precise three-dimensional Fig. 1, no subtraction or image filtering method was applied.
layouts of columnar structures in mature and developing brains,
Analysis of iso-orientation maps. Similar image-processing methods
regardless of their location in the brain. were used to construct the iso-orientation maps based on negative or
positive BOLD data. Analogous to the standard analysis of optical-imag-
METHODS ing data32,33,37, responses to the four different orientations were obtained
Animal preparation. All animal experiments were performed with insti- by dividing each single orientation map (raw percent-change maps) by
tutional approval. Animals were initially anesthetized with a ketamine the normalized sum of four orientations (cocktail blank). For display-
(1025 mg per kg, i.v.) and xylazine (2.5 mg per kg) cocktail. The ani- ing the maps, the dynamic range of the negative (Fig. 2ad) and posi-
mals were intubated and artificially ventilated (2535 strokes per min, tive (Fig. 3a and b) maps were clipped at the upper and lower three
1530 ml per stroke) under isoflurane anesthesia (0.81.5%) in a N2O:O2 percent of the distribution and linearly mapped on an eight-bit gray
mixture of 70:30 throughout the experiment. The animals eyes were scale. In both the negative and positive maps, high-frequency noise was
refracted and corrective contact lenses used if necessary. The animal was removed using a 3 3 pixel Gaussian kernel. The complementarity of
placed in a cradle and restrained in normal postural position using a cus- the orthogonal maps was tested by calculating the linear correlation coef-
tom-designed stereotaxic frame. Endtidal CO2, respiration rate and rec- ficient between the two maps48. Correlation coefficient p-values were
tal temperature were monitored throughout the experiment. Following calculated using standard algorithms48. For the MR time courses in Fig.
2000 Nature America Inc. http://neurosci.nature.com

the three-to-five-hour MR experiments, gas anesthesia was discontin- 2, the ROI for the 45 columns was selected on the basis of negative
ued, and animals were kept on the respirator until they could breathe responses during only the 45 stimulation. The same ROI was then used
independently. The animals were observed for 0.52.0 h before being to plot the time course of those pixels during the 135 stimulation. Like-
returned to their littermates. wise, the ROI for the 135 columns was selected on the basis of negative
responses during only the 135 stimulation. This ROI was then used to
Stimulation protocol. Animals were stimulated binocularly. Visual stim- plot the time course of the pixels during the 45 stimulation. The differ-
uli consisted of high-contrast square-wave moving gratings (0.15 cycles ences in positive signal contrasts during the 45 and 135 stimulations
per degree, 2 cycles per s) of four different orientations (0, 45, 90, were therefore independent of the initial pixel selection process.
135), optimized to elicit responses from neurons in area 18 of the cat
visual cortex29. During the resting period, a stationary grating pattern Analysis of angle maps. Composite angle maps for the negative BOLD
of identical spatial frequency and orientation was presented. A video pro- changes (Fig. 4a) were obtained through a pixel-by-pixel vector addition
jector (Resonance Technology, Northridge, California; resolution, of the four iso-orientation maps (see above) with the negative percent
640 480 pixels) was used to project the visual stimuli onto a screen changes as vector amplitudes and the respective stimulus orientations as
positioned 15 cm from the animals eyes. The screen covered about 37 of vector angles7,32,33. Such composite angle maps were obtained for four
the animals visual field. different experiments that yielded the best contrast-to-noise ratio. The
resulting angle at each pixel was color-coded using a circular color table.
MR methods. After placing the animal in a cradle, we placed a small sur- The positions and densities of the pinwheel centers were detected by cal-
face coil (diameter, 1.2 cm) on top of the animals head corresponding culating the spatial gradients of the composite angle map32,37 and checked
to the Horsley-Clark A3. All MR experiments were performed on an visually. As a control, composite maps were also calculated based on
Oxford (4.7 T, 40 cm in diameter; Oxford, UK) or Magnex (9.4 T, 31 cm MR signals obtained before visual stimulation (Fig. 4b) and those
in diameter; Abingdon, UK) horizontal MRI scanner equipped with a obtained during delayed positive BOLD responses (Fig. 4c; 820 s after
magnetic field gradient of 15 or 30 gauss per cm, respectively. Data stimulus onset). For the sake of consistency, the composite maps in Fig.
obtained at two magnetic field strengths were analyzed separately. The 4ac were generated using identical methods for the region of interest,
amplitude and phase of the biphasic time courses obtained at the two threshold and vector-summation.
field strengths however, were averaged together, as they yielded compa-
rable results. BOLD measurements on a single image slice was made using
gradient-echo echo-planar imaging (EPI). The imaging slice was posi- ACKNOWLEDGEMENTS
tioned 500 m below the pia to avoid superficial draining vessels. The
We thank K. Ugurbil for continuing support of our project and S. Ogawa, A.
MR imaging parameters were data matrix, 64 64; single-shot (4.7 T)
Grinvald, R.B. Tootell, J. Ashe and A. P. Georgopoulos for suggestions and com-
or 2-shot (9.4 T) EPI; FOV, 2 cm 2 cm; slice thickness, 2 mm; TE, 31
ms (4.7 T) or 12 (9.4 T) ms; TR, 0.5 s. A total of 160 images were acquired ments. H. Merkle and J. Strupp provided support in hardware and software.
during each epoch (60 images before stimulation, 20 images during stim- This work was supported by the NIH (NS38295, MH57180, NS10930,
ulation and 80 images after stimulation). Images with a 64 64 matrix RR08079), the Minnesota Medical Foundation and the Keck Foundation.
were zero-filled to a 128 128 matrix, resulting in nominal in-plane res-
olution of 156 156 m2 per pixel. Images obtained for the same orien-
tations within a single fMRI session were averaged for signal-to-noise RECEIVED 20 AUGUST; ACCEPTED 29 NOVEMBER 1999
ratio improvement (usually five to ten epochs).

1. Brodmann, K. Vergleichende Lokalisationslehre der Grosshirnrinde (Johann


Functional image construction. To determine the existence of BOLD Ambrosius Barth, Leipzig, 1909).
activity per se, the MR signals were first cross-correlated with the stimu- 2. Hubel, D. H. & Wiesel, T. N. Receptve field, binocular interactions and
lation protocol47. The cross-correlation coefficient threshold was set at functional architecture in the cats visual cortex. J. Physiol. (Lond.) 160,
> 98% confidence level. The percent-change maps within the negative 106154 (1962).
3. LeVay, S., Stryker, M. P. & Shatz, C. J. Ocular dominance columns and
and positive portions of BOLD time course were averaged into respec- their development in layer IV of cats visual cortex. J. Comp. Neurol. 179,
tive time-binned maps (positive and negative map, respectively). For the 223244 (1978).
map of the negative response, individual pixels showing negative percent 4. LeVay, S. & Nelson, S. B. Vision and Visual Dysfunction 266315
change 0.51.0 s.d. from the mean baseline percent change were taken (Macmillan, Houndsmill, 1991).
5. Sokoloff, L. et al. The 14C-deoxyglucose method for the measurement of
as statistically significant. Because we used clustered pixel analysis with a local cerebral glucose utilization: theory, procedure, and normal values in
cluster size of 4 pixels, the nominal p-value for our threshold was p < 0.2 the conscious and anesthetized albino rat. J. Neurochem. 28, 897916
(ref. 47). The same threshold was used for calculating the four different (1977).

168 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

6. Grinvald, A., Lieke, E., Frostig, R. D., Gilbert, C. D. & Wiesel, T. N. Functional 27. Marota, J. J. A. et al. Investigation of the early response to rat forepaw
architecture of cortex revealed by optical imaging of intrinsic signals. Nature stimulation. Magn. Reson. Med. 41, 247252 (1999).
324, 361364 (1986). 28. Silva, A. C., Lee, S.-P., Iadecola, C. & Kim, S.-G. Early characteristics of CBF
7. Tso, D. Y., Frostig, R. D., Lieke, E. E. & Grinvald, A. Functional organization and deoxyhemoglobin changes during somatosensory stimulation. J. Cereb.
of primate visual cortex revealed by high resolution optical imaging. Science Blood Flow Metab. 20, 201206 (2000).
249, 417420 (1990). 29. Movshon, J. A., Thompson, I. D. & Tolhurst, D. J. Spatial and temporal
8. Frostig, R. D., Lieke, E. E., Tso, D. Y. & Grinvald, A. Cortical functional contrast sensitivity of neurons in areas 17 and 18 of the cats visual cortex. J.
architecture and local coupling between neuronal activity and the Physiol. (Lond.) 283, 101120 (1978).
microcirculation revealed by in vivo high-resolution optical imaging of 30. Loewel, S., Freeman, B. & Singer, W. Topographic organization of the
intrinsic signals. Proc. Natl. Acad. Sci. USA 87, 60826086 (1990). orientation column system in large, flat-mounts of the cat visual cortex: A 2-
9. Bonhoeffer, T. & Grinvald, A. The layout of iso-orientation domains in area deoxyglucose study. J. Comp. Neurol. 255, 401415 (1987).
18 of cat visual cortex: optical imaging reveals a pinwheel-like organization. J. 31. Jezzard, P., Rauschecker, J. P. & Malonek, D. An in vivo model for functional
Neurosci. 13, 41574180 (1993). MRI in cat visual cortex. Magn. Reson. Med. 38, 699705 (1997).
10. Stetter, M. & Obemayer K. Simulation of scanning laser technique for optical 32. Bonhoeffer, T. & Grinvald, A. Iso-orientation domains in cat visual cortex are
imaging of blood-related intrinsic signals. J. Opt. Soc. Am. A 16, 5870 arranged in pinwheel-like pattern. Nature 353, 429432 (1991).
(1999). 33. Kim, D.-S. & Bonhoeffer, T. Reverse occlusion leads to a precise restoration of
11. Ogawa, S. et al. Intrinsic signal changes accompanying sensory stimulation: orientation preference maps in visual cortex. Nature 370, 370372 (1994).
functional brain mapping with magnetic resonance imaging. Proc. Natl. 34. Swindale, N. W, Matsubara, J. A. & Cynader, M. S. Surface organization of
Acad. Sci. USA 89, 59515955 (1992). orientation and direction selectivity in cat area 18. J. Neurosci. 7, 14141427
12. Kwong, K. K. et al. Dynamic magnetic resonance imaging of human brain (1987).
activity during primary sensory stimulation. Proc. Natl. Acad. Sci. USA 89, 35. Diao, Y. C., Jia, W. G., Swindale, N. V. & Cynader, M. S. Functional
56755679 (1992). organization of the cortical 17 per 18 border region in the cat. Exp. Brain Res.
13. Loewel, S. & Singer, W. Tangential intracortical pathways and the 79, 271282 (1990).
development of iso-orientation bands in cat striate cortex. Dev. Brain Res. 56, 36. Maldonado, P. E., Goedecke, I., Gray, C. M. & Bonhoeffer, T. Orientation
2000 Nature America Inc. http://neurosci.nature.com

99115 (1990). selectivity in pinwheel centers in cat visual cortex. Science 276, 15511555
14. Fox, P. T. & Raichle, M. E. Focal physiological uncoupling of cerebral blood (1997).
flow and oxidative metabolism during somatosensory stimulation in human 37. Shmuel, A. & Grinvald, A. Functional organization for direction of motion
subjects. Proc. Natl. Acad. Sci. USA 83, 11401144 (1986). and its relationship to orientation maps in cat area 18. J. Neurosci. 16,
15. Posner, M. I. & Raichle, M. E. Images of Mind (Freeman, New York, 1994). 69456964 (1996).
16. Wagner, A. D. et al. Building memories: remembering and forgetting of verbal 38. Crair, M. C., Gillespie, D. C. & Stryker, M. P. The role of visual experience in
experiences as predicted by brain activity. Science 281, 11881191 (1998). the development of columns in cat visual cortex. Science 279, 566570 (1998).
17. Kim, S.-G. et al. Functional magnetic resonance imaging of motor cortex: 39. Rojer, A. S. & Schwartz, E. L. Cat and monkey cortical columnar patterns
hemispheric asymmetry and handedness. Science 261, 615617 (1993). modeled by bandpass-filtered 2D white noise. Biol. Cybern. 62, 381391
18. DeYoe, E. A. et al. Mapping striate and extrastriate visual areas in human (1990).
cerebral cortex. Proc. Natl. Acad. Sci. USA 93, 23822386 (1996). 40. Blasdel, G. G. Differential imaging of ocular dominance and orientation
19. Sereno, M. I. et al. Borders of multiple visual areas in humans revealed by selectivity in monkey striate cortex. J. Neurosci. 12, 31153138 (1992).
functional magnetic resonance imaging. Science 268, 889893 (1995). 41. Menon, R. S., Ogawa, S. Strupp, J. P. & Ugurbil, K. Ocular dominance in
20. Tootell, R. B. et al. Functional analysis of V3A and related areas in monkey human V1 demonstrated by functional magnetic resonance imaging. J.
visual cortex. J. Neurosci. 17, 70707078 (1997). Neurophysiol. 77, 27802787 (1997).
21. Engel, A. S., Glover, G. H. & Wandell, B. A. Retinotopic organization in 42. Ogawa, S., Menon, R. S., Kim, S.-G. & Ugurbil, K. On the characterizatics of
human visual cortex and the spatial precision of functional MRI. Cereb. functional magnetic resonance imaging of the brain. Annu. Rev. Biophys.
Cortex 7, 181192 (1997). Biomol. Struct. 27, 447474 (1998).
22. Malonek, D. & Grinvald, A. Interactions between electrical activity and 43. Janz, C., Speck, O. & Henning, J. Time-resolved measurements of brain
cortical microcirculation revealed by imaging spectroscopy: Implication for activation after a short visual stimulus: new results on the physiological
functional brain mapping. Science 272, 551554 (1996). mechanisms of the cortical response. NMR Biomed. 10, 222229 (1997).
23. Malonek, D. et al. Vascular imprints of neuronal activity: relationships 44. Vanzetta, I. & Grinvald, A. Increased cortical oxidative metabolism due to
between the dynamics of cortical blood flow, oxygenation, and volume sensory stimulation: implications for functional brain imaging. Science 286,
changes following sensory stimulation. Proc. Natl. Acad. Sci. USA 94, 15551558 (1999).
1482614831 (1997). 45. Yacoub, E. & Hu, X. Detection of the early negative response in fMRI at 1.5
24. Ernst, T. & Henning, J. Observation of a fast response in functional MR. Tesla. Magn. Reson. Med. 41, 10881092 (1999).
Magn. Reson. Med. 32, 146149 (1994). 46. Logothetis, N. K., Guggenberger, H., Peled, S. & Pauls, J. Functional imaging
25. Menon, R. S. et al. BOLD based functional MRI at 4 Tesla includes a capillary of the monkey brain. Nat. Neurosci. 2, 555562 (1999).
bed contribution: Echo planar imaging correlates with previous optical 47. Forman, S. D. et al. Improved assessment of significant activation in
imaging using intrinsic signals. Magn. Reson. Med. 33, 453459 (1995). functional magnetic resonance imaging (fMRI): use of a cluster-size
26. Hu, X., Le, T. H. & Ugurbil, K. Evaluation of the early response in fMRI in threshold. Magn. Reson. Med. 33, 636647 (1995).
individual subjects using short stimulus duration. Magn. Reson. Med. 37, 48. Press, W. H., Teukolsky, S. A., Vetterling, W. T. & Flannery, B.P. Numerical
877884 (1997). Recipes in C (Cambridge, UK, 1992).

nature neuroscience volume 3 no 2 february 2000 169


2000 Nature America Inc. http://neurosci.nature.com

articles

Dynamics of perceptual oscillations


in form vision
Hugh R. Wilson1, Boris Krupa1 and Frances Wilkinson2

1 Visual Science Center, University of Chicago, 939 E. 57th Street, Chicago, Illinois 60637, USA
2 Department of Psychology, 1205 Docteur Penfield Avenue, McGill University, Montreal, Quebec, H3A 1B1, Canada
Correspondence should be addressed to H.R.W. (hrw6@midway.uchicago.edu)

Certain periodic dot patterns (Marroquin patterns) generate a percept of dynamically oscillating
circles, and analogous effects were explored by op artists in the 1960s. Here we show psychophysically
that circles are perceived in these patterns only around specific points that are quantitatively predicted
2000 Nature America Inc. http://neurosci.nature.com

by a neural model of configural units hypothesized to reside in cortical area V4. Circles superimposed
on the pattern mask perception of illusory circles. A neural model of lateral inhibitory interactions
among V4 configural units showing spike-frequency adaptation quantitatively accounts for the human
data. The model is consistent with ideas on the neural basis of attention in V4, and it suggests that
attention may be biased via neuromodulation of slow hyperpolarizing potentials in cortical neurons.

Certain static patterns generate dynamic visual percepts, an extract the center and mean radius of a face transparently added
effect on which op artists capitalized frequently during the to a house (Fig. 1b), a stimulus that has been used to provide
1960s1,2. One salient example is the pattern (Fig. 1a) created in evidence for object-based attention17. Thus, V4 concentric units
1976 by Marroquin3. This pattern is produced by superimpos- may be important in face and other ellipsoidal object analysis as
ing three copies of a square dot grid, each copy being rotated well as in selective attention.
by 60 relative to the others. This static pattern generates a per- Selective attention effects are evident in V4 at the single neu-
cept of circular shapes that appear and vanish at various loca- ron level1821 and are believed to result from inhibitory compe-
tions in an oscillatory fashion. Vision scientists have tition in parallel networks22. This suggested that the Marroquin
characterized this and related oscillating patterns as products illusion might arise from competitive interactions among V4
of dynamic grouping4,5. Such explanations, however, raise ques- concentric units, and that the illusion might therefore provide
tions concerning the nature of this grouping and the underlying further insight into the mechanisms of selective attention in V4.
dynamical processes producing oscillations. Here we apply psy- The results reported here confirm that inhibitory interactions
chophysical techniques to measure the frequency and duration among model V4 concentric units account quantitatively for vis-
of circle visibility throughout the Marroquin pattern, and we ibility of Marroquin circles throughout the pattern. The resul-
demonstrate that a plausible neural model can quantitatively tant neural network may therefore form a basis for spatial or
account for the data. object based selective attention, with neuromodulation of spike-
In searching for a possible neural basis for the oscillating cir- frequency adaptation providing a mechanism for top-down bias-
cles in the Marroquin pattern, a connection with the perception ing of attentional effects.
of circular structure in Glass patterns6,7 suggested itself. (See refs.
8, 9 for examples of Glass patterns.) Circular structure in these RESULTS
random-dot patterns is considerably more salient than radial, Psychophysics
hyperbolic or translational structure8,9. The visual system extracts To understand the dynamics underlying the Marroquin illusion,
concentric structure from Glass patterns by linear pooling of we first asked whether circles were more likely to appear at some
quasi-concentric orientations, and a neural model derived from locations than at others. The visibility of circles centered at dif-
these observations provides a quantitative explanation of the ferent points in the Marroquin pattern was quantified using a
data8. Comparison with primate single-unit physiology1012 sug- procedure analogous to that generally used in binocular rivalry
gests that such concentric units might arise in V4, an intermedi- measurements23. In each experiment, subjects fixated a red dot at
ate area in the ventral form-vision system13,14. Evidence for units a designated position within the Marroquin pattern and depressed
optimized for analysis of quasi-concentric structure in human a button whenever an illusory circle was perceived to be centered
V4 is supported by converging evidence from fMRI (James et al., on that dot (see Methods). Because of the hexagonal symmetry
Invest. Ophthalmol. Vis. Sci. 40, S819, Abstr. 4313, 1999), event- of these patterns, measurements were restricted to ten dot posi-
related potentials recorded directly from the human cortical sur- tions within one sextant of the pattern. Visibility was defined as
face15 and studies of a patient with unilateral V4 damage (Mazer the fraction of a two-minute viewing period during which an illu-
et al., Soc. Neurosci. Abstr. 25, 212.14, 1999). Neural simulations sory circle of specified diameter (line in Fig. 1a) was perceived to
have demonstrated that model V4 units can provide a popula- surround the designated center dot. Data for one subject are plot-
tion code for the location, shape and bilateral symmetry of ted at all ten positions for runs on three different days (Fig. 2a).
human heads16. For example, model V4 concentric units can Although data at different positions were collected in pseudo-

170 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 1. Patterns generating percepts of circular or ellipsoidal structure.


a (a) Marroquin pattern3 produced by superimposing three square grids
of dots, each rotated by 60 relative to the others. In experiments, dots
were presented against a gray background of mean luminance 50.5 cd
per m2. While subjects fixate this pattern, illusory circles appear and
vanish centered at various locations throughout the pattern. Although
illusory circles of several different diameters are sometimes seen, with
the dot separations and viewing distances used in our experiments, only
illusory circles of one size were apparent (diameter indicated by hori-
zontal line in lower right). (b) Simulated response of model V4 concen-
tric units to a transparent face/house image of the type used in a study of
object-based attention17. Processing of the face alone produced a maxi-
mum model response at the point indicated by the upper black circle.
The mean radius of the stimulus was encoded by the maximum model
response and corresponded to the distance indicated by the arrow. The
model population response also encoded the stimulus as vertically elon-
gated16. Superposition of the house onto the face only shifted the maxi-
mum V4 model response to the lower black circle but left both the
estimated radius and axis of elongation essentially unchanged. Model V4
concentric units only responded to the face shape and thus are capable
2000 Nature America Inc. http://neurosci.nature.com

b of extracting ellipsoidal object boundaries from complex images.

(Fig. 3a). These masking circles were centered at varying dis-


tances from the illusory center circle in different experiments.
Both large circles (diameter 1.80) and smaller circles (diameter
0.90) produced significant reductions in the visibility of the illu-
sory circle centered about position 1 (Fig. 3b). Significantly,
masking data from both circle sizes fell off as the same function
F of increasing radius R from position 1:
F(R) = 1 A exp ( R5/5) (1)
Only the amplitude A increased with increasing circle diam-
eter, and s = 1.97 for both diameters. This suggests lateral
inhibitory interactions among units optimized to extract quasi-
circular structure from stimuli. To test this idea further, the cir-
cular masks were replaced with four parallel vertical lines having
the same separations as the circle diameter and the same total
length as the circle circumferences. These lines produced almost
no masking of illusory circle visibility (Fig. 3b). This provides
random order on different days, performance was highly repro- further evidence that Marroquin patterns evoke lateral inhibi-
ducible from day to day. Results averaged over three days of exper- tion among units selective for concentric structure.
iments for each of four subjects show a similar pattern (Fig. 2b).
In comparing subjects, visibilities were normalized relative to that Neural model
at position 1. (Absolute visibilities are listed in Table 1.) Although The Marroquin oscillations can be explained by extending a neur-
there was some variability among subjects, illusory-circle visibil- al model developed to account for the visibility of concentric
ity was high for all subjects at positions 1, 7 and 10, whereas vis- structure in Glass patterns8. This model uses a sequence of ori-
ibility was low at positions 5 and 8. One-way ANOVA showed a ented filtering, full-wave rectification and orthogonal second-
significant effect of position (F9,110 = 8.42, p < 0.0001). A post-hoc stage oriented filtering (Fig. 5) to extract local curvature
Fischers PLSD showed significant differences between each of information from the stimulus25,26. There is direct psychophysi-
positions 1, 7 and 10 and positions 5 and 8 at p < 0.0001.) Clear- cal evidence that the final stage incorporates linear concentric
ly, therefore, Marroquin patterns generate illusory circles prefer- pooling of curvature information8,9. Responses of such model
entially surrounding particular hot spots for all subjects, whereas units agree with a subset of neurons recorded in V4 in several
circles are rarely perceived around other cold spots. studies1012, and we will therefore refer to them as model V4 con-
Mean durations of illusory circle visibility are indicated for centric units. Processing the Marroquin stimulus by an array of
position 1 (the center of the pattern) in Table 1. Two subjects these units produced a response showing strong activation of
(FW and HRW) showed mean durations in the 23.5 s range, model V4 units by certain pattern locations, but little or no acti-
similar to binocular rivalry23,24, and both produced standard devi- vation by other locations (Fig. 4a). The three hot spots and two
ations comparable to the mean. The remaining two subjects had cold spots from the psychophysical data are marked, demon-
considerably longer mean visibilities. strating qualitative agreement of the model responses with the
As the Marroquin oscillations are suggestive of instabilities in data. A quantitative comparison among all ten positions demon-
a competitive network, we wondered whether pattern masking strates that the model responses agree well with relative visibili-
might be effective in altering the mean visibility of illusory cir- ties averaged across subjects (Fig. 2c). Thus, the V4 concentric
cles. To test this, further data were gathered at position 1 with a model accurately predicts relative visibilities of illusory circles at
superimposed mask comprising four surrounding gray circles different locations throughout the Marroquin pattern.

nature neuroscience volume 3 no 2 february 2000 171


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 2. Visibility of illusory circles centered at ten different points in the


a Marroquin pattern. Because of hexagonal symmetry, these points were
FW #1 all chosen from one sextant of the pattern. Position 1 was the center of
FW #2 the pattern, and successively higher numbered points progressed out-
FW #3 ward toward the upper left. (a) Visibility (fraction of time an illusory cir-
cle was seen during a two-minute run) is plotted for all positions from
three different days of experiments on FW. Despite pseudorandom
Visibility

ordering of data collection from day to day, a high degree of repeatabil-


ity is evident. (b) Mean data for all four subjects are plotted normalized
to the relative visibility at position 1. (Absolute visibilities are listed in
Table 1.) Illusory circles were highly visible at positions 1, 7 and 10 for
all subjects, whereas circles were seldom visible around positions 5 and
8. These five positions have been marked (Fig. 4a). This dependence of
illusory circle visibility was highly statistically significant (see text). (c) V4
model predictions of visibility variations across positions are compared
with data averaged across all subjects.
Position
b
Designating excitatory and inhibitory neuron spike rates by
2000 Nature America Inc. http://neurosci.nature.com

E and I, and the AHP current by H, a suitable set of equations to


describe competitive interactions within a two-dimensional array
Relative visibility

of model V4 concentric cells is


2
dEn 100 P+
E = En + 2 2
dt (10 + Hn) + P+

where P = SMarroquin 0.6n=k Ik exp ( Rnk5/5)

dIn
I = In + En
dt

dHn
c Position H
dt
= Hn + gEn
(2)
Mean data The first equation here indicates that the excitatory firing rates
Relative visibility or model response

V4 model follow a Naka-Rushton31 function of the net postsynaptic input P


produced by stimulus SMarroquin, extracted from the Marroquin
pattern by the model V4 circuit (Fig. 5a) minus network inhibi-
tion. Naka-Rushton functions provide good fits to firing rates of
visual cortical neurons32,33. Each excitatory neuron En activates
an inhibitory neuron In, which in turn provides subtractive inhi-
bition to a range of neighboring E neurons (excluding En) using
the connectivity function of relative distance Rnk derived from
equation (1). Finally, adapting effects of the AHP current are sim-
ulated by an increase in the semi-saturation constant of the Naka-
Rushton function mediated by Hn, a formulation that provides
a reasonable approximation to cortical neuron adaptation27. In
Position
general, g = 2.9 (but it is assumed to be under neuromodulatory
control; see Discussion), and the time constants were assigned
The oscillations of illusory circles in the Marroquin pattern the plausible values of E = 15 ms, I = 8 ms and H = 400 ms.
must now be explained. The psychophysical masking data pro- These constants reflect the faster spiking of I than of E neurons28,
vide evidence for lateral inhibitory interactions that decline with and H falls within the range reported for AHP currents in human
the distance between concentric units, and this suggests the exis- cortical neurons30,34.
tence of a spatially regional winner-take-all (WTA) network The temporal response from a model E neuron at position 1
among V4 concentric units. WTA networks in general, however, of the Marroquin pattern shows the effects of spike-frequency
cannot generate oscillations unless the winners adapt or fatigue adaptation in the rapid drop of spike rate to about one-third of
with time27. Here a property of virtually all neocortical excitatory the initial transient rate (Fig. 6a). As this neuron continues to
(but not inhibitory) neurons in humans and other mammals sug- fatigue further during its firing plateau, the effects of time-varying
gests itself, namely, spike-frequency adaptation2830. When stim- inhibitory inputs become evident. Ultimately, inhibition termi-
ulated by a constant-current input, neocortical excitatory neurons nates spiking in this neuron, and neighboring neurons become
initiate firing at a high rate that typically drops by a factor of about active. Repeated simulations of the two-minute duration of our
3.0 within a few hundred ms. This spike-frequency adaptation is experiments with slightly different initial conditions produced
mediated by a slow Ca2+-mediated K+ afterhyperpolarizing cur- the model responses summarized in Table 1. Similar to FW and
rent (AHP). Inclusion of spike-frequency adaptation in a region- HRW, the model predicted a visibility at position 1 of 0.62 and a
al WTA network of model V4 concentric units produces a mean visibility duration of 2.24 1.93 s. Durations for both the
quantitative account of Marroquin pattern oscillations. model and human subjects were plotted in a histogram and fit-

172 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a b

Relative visibility
1.8 circle
0.9 circle
lines
2000 Nature America Inc. http://neurosci.nature.com

Mask displacement

Fig. 3. Masking of illusory circle visibility. (a) The masking stimulus comprised four circular contours superimposed on the Marroquin pattern. These mask-
ing circles were always spaced at equal distances from position 1, with distance being varied from experiment to experiment. As shown by mean data for
four subjects (b), the masking circles greatly reduced the relative visibility of illusory circles centered on position 1, with larger effects being produced by
larger diameter masking circles. Data for both circle sizes were well fit by the function in equation (1), with only amplitude A depending on size. For com-
parison, four vertical parallel lines with the same total length as the masking circle circumferences and spaced apart by a distance equal to the circle diame-
ters produced almost no noticeable masking. These data thus reflect a form of pattern-specific masking rather than simple contour interactions per se.

ted with a gamma distribution function (Fig. 6b and c). The radial gratings than by conventional sinusoidal gratings,
model and subjects both show comparably good fits to the gamma although these studies did not test for the possibility of com-
distribution (parameters summarized in Table 1). To illustrate the petitive interactions.
overall spatial percept implied by the model, neural responses The complex oscillations in this regional winner-take-all net-
30 seconds into one simulation were convolved with the activating work are driven by spike-frequency adaptation in excitatory neu-
input extracted from the Marroquin pattern, which produced a rons, which is known to be caused by AHP currents 2830 .
circular configuration resembling the percept (Fig. 4b). Physiological studies of excitatory neurons from human neo-
As the model responses were sufficiently complex to suggest cortex have shown that AHP currents are modulated by hista-
chaos, further simulations were conducted to compute the max- mine, acetylcholine, norepinephrine and serotonin, all of which
imum Lyapunov exponent using standard techniques27. The reduce the magnitude of spike-frequency adaptation34. Effects of
extreme sensitivity to initial conditions of a chaotic system is such neuromodulation in the model V4 competitive network can
manifested by a positive . We found that = 0.10/ms, so the be incorporated simply by reducing the parameter g in equation
neural model is not chaotic but rather represents a highly com- (2). Simulations have shown that a 14% reduction to g = 2.5 caus-
plex, asymptotically stable limit cycle oscillation. Rather than es the model to produce a 0.92 visibility as demonstrated by sub-
representing a simple alternation between two states as in binoc- ject EL (Table 1). This provides a plausible explanation for
ular rivalry, this oscillation cycles through a very large ensemble individual differences in our study.
of states before repeating. As discussed elsewhere8,9, the filterrectifyfilter sequence at
the earlier stages of the model is incorporated into many models
DISCUSSION for texture segregation. The unique aspect of our model is the
The foregoing analysis demonstrates that the Marroquin illu- final concentric, linear summation stage, which was derived from
sion can be explained quantitatively by inhibitory interactions psychophysics8. To date, proponents of other approaches to texture
giving rise to a regional winner-take-all
computation. Input to this network is Table 1: Summary of data for four subjects and V4 model simulation.
provided by model units that explain
Subject Visibility Duration (s) Gamma n Gamma (1/s)
the perception of structure in circular
FW 0.57 2.02 2.57 1.59 2.45
Glass patterns 8 . As these units have
properties similar to a subset of primate HRW 0.62 3.56 2.92 2.01 1.00
V4 neurons1012, it is hypothesized that BK 0.77 6.33 4.24 1.33 0.46
the competitive neural network devel- EL 0.91 32.67 46.78 n/a n/a
oped here reflects human V4 activity8,9. V4 Model 0.62 2.24 1.93 1.72 1.56
Results from fMRI (James et al., Invest.
Mean visibilities and mean durations are reported for position 1 (center of pattern). The two gamma para-
Ophthalmol. Vis. Sci. 40, S819, Abstr. meters are from least mean squares fits of K (t)n exp (t) to histograms of the duration data (see Fig.
4313, 1999) and event-related poten- 6). No gamma parameters could be calculated for EL because of the very long durations and consequent
tials15 indicate that human V4v is more small number of duration data. Model results fall within the data range for the first three subjects, and
agreement with EL can be obtained by a small shift in one model parameter (see text).
strongly activated by concentric and

nature neuroscience volume 3 no 2 february 2000 173


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 4. Pseudocolor representa-


tion of model V4 concentric unit8
responses to the Marroquin pat- a b
tern. (a) From maximum to zero,
responses are plotted in white,
yellow, red, dark red and black. 10
Model responses at the three high 8
points (positions 1, 7 and 10) and
two low points (positions 5 and 8) 1
of the human data are marked.
Relative model responses at all 7
ten positions agree well with 5
mean human data (Fig. 2c).
(b) One frame of the neural net-
work simulation in equation (2).
The small cross-shaped patches
are active local islands of E neu-
rons that have suppressed all
other neural activity in their vicin-
ity. Illusory circles have been
2000 Nature America Inc. http://neurosci.nature.com

added by convolving the network


activity pattern with a circle representing the effective input to these neurons from the Marroquin patterns. The circles thus represent the percept sig-
naled by network activity. (An animation of V4 network dynamics may be viewed on the web at http://neurosci.nature.com/web_specials/)

grouping have not attempted to explain the Marroquin illusion. It Modifications of the current model may therefore be useful in
is not clear how such a model might operate, but it will be inter- explaining the patchwork percept and spreading waves in
esting to see whether alternative approaches can provide a com- binocular rivalry.
parable fit to our psychophysical data. In any event, it will still be There have been several theoretical proposals that spatial atten-
necessary to incorporate regional inhibitory interactions and adap- tion may reflect the activity of winner-take-all networks40,41. Fur-
tation to account for the observed perceptual oscillations. thermore, studies of primate V4 demonstrate prominent
Both the data and model reported here show similarities to attentional effects1821,4244. This work has led to the proposal that
binocular rivalry. The Marroquin illusion and rivalry both gen- selective attention results from biasing of competitive interactions
erate visibility intervals well approximated by a gamma distri- in parallel networks22. It is therefore reasonable to hypothesize that
bution 23,24,35 . In addition, rivalry has been modeled by the parallel competitive network embodied in equation (2) reflects
competitive interactions plus fatigue36, and there is evidence one substrate for selective attention operating among V4 units. If
that rivalry is not chaotic37,38. Finally, it has been shown that so, a novel form of network biasing suggests itself: localized reduc-
disappearances such as those of the Marroquin circles are sim- tion of spike-frequency adaptation controlled by modulatory neu-
ilar to binocular rivalry and are not caused by eye movements39. rotransmitters. Changes in the magnitude of spike-frequency

3D model Model cross section Fig. 5. Schematic of the complete V4 concen-


tric unit dynamical model illustrated both in
perspective (left) and in cross-section (right)
for clarity. V1 filtering was performed at 12
different simple cell orientations spaced at
15 intervals. The filtered response at each
orientation was then full-wave rectified and
filtered by a larger second-stage oriented fil-
ter (hypothesized to reside in V2) at an
orthogonal orientation, a sequence of opera-
tions that extracts curvature information27,28.
Finally, responses of 24 concentrically
arranged second-stage filters are summed lin-
early (S in the model cross section) to pro-
duce the input to model V4 concentric units.
Further details of this feedforward model for
concentric V4 units were reported previ-
ously8,9. At the V4 stage, these units engage in
competitive lateral inhibition (lines with solid
circles at the top of the diagram) over spatial
regions defined by the masking function in
equation (1). Given the dimensions of the
connectivity function, each neuron inhibited a
circular region comprising approximately 12%
(at 1/e level) of the 64 64 network. There
was no inhibition between a unit and itself or
its two nearest neighbors in each direction.

174 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 6. Comparison of model net


work responses and human data. a
(a) Temporal response of the model E Transient
neuron in position 1 (center of the
network) is plotted. Following a tran-

Spike rate
sient overshoot when the neuron
escapes from inhibition and switches Inhibition
on, the rapid drop of the spike rate to
about 1/3 of the initial peak value
results from spike-frequency adapta-
tion mediated by H in equation (2).
Small fluctuations in firing level during
the subsequent plateau result from
time-varying inhibition arising from
oscillations elsewhere in the network. Time (s)
Ultimately, competition and spike-fre-
quency adaptation cause the neuron b
to cease firing. Histograms of visibility FW Model
durations at position 1 are shown for Gamma Gamma
both subject FW (b) and the V4 model n = 103
2000 Nature America Inc. http://neurosci.nature.com

n = 478
(c). Both are well fit by a distribution

Interval count
Interval count

(parameters in Table 1).

adaptation would have a small


effect on the initial transient
response of cortical neurons, but
would significantly enhance the
subsequent asymptotic firing level; Interval duration (s) Interval duration (s)
this time course of adaptational
effects is observed in primate V4
neurons21. Aspects of selective attention in visual areas V1 and V2 series of visibility durations was obtained. Each experiment was cen-
might be explained by incorporating recurrent connections from tered at one of ten points in the Marroquin pattern, and each experi-
the V4 model to these areas, thus generating a competitive atten- ment lasted two min. To minimize fading of dots due to retinal
tional hierarchy41. Delayed figureground enhancements observed adaptation, the dots were counterphase flickered between black and
in V1 might also be explained by such feedback to V1 from V4 white at 1.5 Hz, which made the task considerably easier for subjects.
concentric units45. Data were collected at all fixation points on each of three days, with
Despite their intrinsic fascination, illusions attain their pri- points measured in pseudo-random order each day.
mary significance as windows for glimpsing underlying visual
processes. From this perspective, the Marroquin illusion may be Modeling. Simulations of equation (2) were conducted in MatLab
software on a Macintosh G3 computer using a Runge-Kutta routine
viewed as the result of presenting a dense set of competing stim-
with constant step size. Before solving the differential equations, we
uli to an attentional network. Similarly, the oscillation triggered applied the concentric V4 unit model8 consisting of oriented V1 fil-
by this set of stimuli may be construed as a form of neural search tering, full-wave rectification, orthogonal V2 filtering and final con-
sequence incorporating inhibition of return46,47 mediated by spike- centric linear pooling to a 512 512 portion of the Marroquin pattern.
frequency adaptation. We have presented evidence that model V4 To reduce the size of the simulation, this filtered input to V4 concen-
concentric units can detect ellipsoidal objects in complex scenes tric units was subsampled down to 64 64. This was carefully done
(Fig. 1b) and that such units interact via spatially regional win- in order to retain the symmetries in the V4 processed image
ner-take-all competition. Given evidence for object-based atten- (see Fig. 4a). Equation (2) therefore represents a system of 3 642 or
17
tion , this raises the exciting possibility that further study of V4 12,288 coupled nonlinear differential equations. The first-stage filters
concentric units may help to elucidate the physiological bases of were derived from earlier masking studies with parameters for 16.0
cycles per degree48. The second-stage filters were scaled for the first-
such attentional processes. stage filters (16.0 cycles per degree) from values previously reported8.
FFTs were used to evaluate the summation in the definition of p in
METHODS equation (2), so boundary conditions were periodic. As p represents
Psychophysics. Marroquin patterns were generated by superimposing the postsynaptic input to excitatory neurons, which are described by
three square dot grids rotated by 60 with respect to each other. At the typical spike rate equations27, p provides an input to the Naka-Rush-
viewing distance of 1.5 m, each dot subtended 40.6 arc s, and the spac- ton function31 that is positive or zero: p+ = max (p, 0). This thresh-
ing between the dots in the square grids was 10.8 arc min. Overall, the olding ensured that negative postsynaptic potentials produced a zero
Marroquin pattern subtended 8.12 8.12. The dots were at 100% con- firing rate. The maximum rate (100) and semi-saturation (10) of the
trast and superimposed on a background of mean luminance 50.5 cd Naka-Rushton function were chosen to produce oscillations in the net-
per m2. In each experiment, three collinear dots were colored red: a cen- work given the strength of the filtered V4 model inputs. The equation
tral one that was fixated by the subject and two others located on the for the I neurons is linear for mathematical convenience; an I equa-
diameter of an illusory circle (see line in Fig. 1a) that the subject was tion with a Naka-Rushton rate nonlinearity and suitable parameters
instructed to monitor. The subjects task was to fixate the center red dot produces very similar results. Because of the wide range of time con-
and depress the mouse button whenever a concentric, illusory circle was stants (8400 ms), these are stiff equations, so a series of preliminary
perceived to pass through the two flanking red dots. By recording the simulations were run to determine an appropriate time step for the
times at which the mouse button was depressed and released, a time simulations, which turned out to be 0.05 ms. The maximum Lyapunov

nature neuroscience volume 3 no 2 february 2000 175


2000 Nature America Inc. http://neurosci.nature.com

articles

exponent was computed by comparing the main trajectory at each 21. McAdams, C. J. & Maunsell, J. H. R. Effects of attention on orientation tuning
time step with a second that started the interval displaced a distance functions of single neurons in macaque cortical area V4. J. Neurosci. 19, 431441
(1999).
of 106 in the state space of the system. This was repeated throughout 22. Desimone, R. & Duncan, J. Neural mechanisms of selective visual attention.
the entire 2.0 min of each simulation, and the maximum Lyapunov Annu. Rev. Neurosci. 18, 193222 (1995).
exponent was then estimated using standard formulas27. 23. Fox, R. & Herrmann, J. Stochastic properties of binocular rivalry alternations.
Percept. Psychophys. 2, 432436 (1967).
Note: An animation of V4 network dynamics can be found on the Nature 24. Blake, R. A neural theory of binocular rivalry. Psychol. Rev. 96, 145167 (1989).
25. Dobbins, A., Zucker, S. W. & Cynader, M. S. Endstopped neurons in the visual
Neuroscience web site (http://neurosci.nature.com/web_specials/). cortex as a substrate for calculating curvature. Nature 329, 438441 (1987).
26. Wilson, H. R. & Richards, W. A. Curvature and separation discrimination at
ACKNOWLEDGEMENTS texture boundaries. J. Opt. Soc. Am. A 9, 16531662 (1992).
27. Wilson, H. R. Spikes, Decisions, and Actions: Dynamical Foundations of
This research was supported in part by NIH grant #EY02158 to H.R.W., by a Neuroscience (Oxford Univ. Press, Oxford, 1999).
grant from Research to Prevent Blindness to the University of Chicago and by 28. Connors, B. W. & Gutnick, M. J. Intrinsic firing patterns of diverse
NSERC grant #OGP0007551 (Canada) to F.W. neocortical neurons. Trends Neurosci. 13, 99104 (1990).
29. Foehring, R. C., Lorenzon, N. M., Herron, P. & Wilson, C. J. Correlation of
physiologically and morphologically identified neuronal types in human
RECEIVED 23 AUGUST; ACCEPTED 3 DECEMBER 1999 association cortex in vitro. J. Neurophysiol. 66, 18251837 (1991).
30. Lorenzon, N. M. & Foehring, R. C. Relationship between repetitive firing and
afterhyperpolarizations in human neocortical neurons. J. Neurophysiol. 67,
1. Barrett, C. Op Art (Studio Vista, London, 1970). 350363 (1992).
2. Wade, N. J. Op art and visual perception. Perception 7, 2146 (1977). 31. Naka, K. I. & Rushton, W. A. S-potentials from colour units in the retina of
3. Marroquin, J. L. Human Visual Perception of Structure. Masters thesis, MIT fish. J. Physiol. (Lond.) 185, 584599 (1966).
2000 Nature America Inc. http://neurosci.nature.com

(1976). 32. Albrecht, D. G. & Hamilton, D. B. Striate cortex of monkey and cat: contrast
4. Marr, D. Vision: A Computational Investigation into the Human response function. J. Neurophysiol. 48, 217237 (1982).
Representation and Processing of Visual Information 4950 (Freeman, San 33. Sclar, G., Maunsell, J. H. R. & Lennie, P. Coding of image contrast in central
Francisco, 1982). visual pathways of the macaque monkey. Vision Res. 30, 110 (1990).
5. Stevens, K. A. & Brookes, A. Detecting structure by symbolic constructions 34. McCormick, D. A. & Williamson, A. Convergence and divergence of
on tokens. Comput. Graph. Image Process. 37, 238260 (1987). neurotransmitter action in human cerebral cortex. Proc. Natl. Acad. Sci. USA
6. Glass, L. Moir effect from random dots. Nature 223, 578580 (1969). 86, 80988102 (1989).
7. Glass, L. & Prez, R. Perception of random dot interference patterns. Nature 35. Borsellino, A., De Marco, A., Allazetta, A., Rinesi, S. & Bartolini, B. Reversal
246, 360362 (1973). time distribution in the perception of visual ambiguous stimuli. Kybernetik
8. Wilson, H. R., Wilkinson, F. & Asaad, W. Concentric orientation summation 10, 139144 (1972).
in human form vision. Vision Res. 37, 23252330 (1997). 36. Lehky, S. An astable multivibrator model of binocular rivalry. Perception 17,
9. Wilson, H. R. & Wilkinson, F. Detection of global structure in Glass patterns: 215228 (1988).
implications for form vision. Vision Res. 38, 29332947 (1998). 37. Richards, W., Wilson, H. R. & Sommer, M. A. Chaos in percepts? Biol.
10. Gallant, J. L., Braun, J. & Van Essen, D. C. Selectivity for polar, hyperbolic, Cybern. 70, 345349 (1994).
and Cartesian gratings in macaque visual cortex. Science 259, 100103 38. Lehky, S. R. Binocular rivalry is not chaotic. Proc. R. Soc. Lond. B Biol. Sci.
(1993). 259, 7176 (1995).
11. Gallant, J. L., Connor, C. E., Rakshit, S., Lewis, J. W. & Van Essen, D. C. Neural 39. Wade, N. J. Distortions and disappearances of geometrical patterns.
responses to polar, hyperbolic, and Cartesian gratings in area V4 of the macaque Perception 6, 407433 (1977).
monkey. J. Neurophysiol. 76, 27182739 (1996). 40. Koch, C. & Ullman, S. Shifts in selective visual attention: towards the
12. Kobatake, E. & Tanaka, K. Neuronal selectivities to complex object features in underlying neural circuitry. Hum. Neurobiol. 4, 219227 (1985).
the ventral visual pathway of the macaque cerebral cortex. J. Neurophysiol. 71, 41. Tsotsos, J. K. Analyzing vision at the complexity level. Behav. Brain Sci. 13,
856867 (1994). 423469 (1990).
13. Mishkin, M., Ungerleider, L. G. & Macko, K. A. Object vision and spatial vision: 42. Motter, B. C. Focal attention produces spatially selective processing in visual
two cortical pathways. Trends Neurosci. 6, 414417 (1983). cortical areas V1, V2, and V4 in the presence of competing stimuli.
14. Van Essen, D. C., Anderson, C. H. & Felleman, D. J. Information processing in J. Neurophysiol. 70, 909919 (1993).
the primate visual system: an integrated systems perspective. Science 255, 43. De Weerd, P., Peralta, M. R., Desimone, R. & Ungerleider, L. G. Loss of
419423 (1992). attentional stimulus selection after extrastriate cortical lesions in macaques.
15. Allison, T., Puce, A., Spencer, D. D. & McCarthy, G. Electrophysiological studies Nat. Neurosci. 2, 753758 (1999).
of human face perception. I: Potentials generated in ocippitotemporal cortex by 44. Reynolds, J. H., Chelazzi, L. & Desimone, R. Competitive mechanisms
face and non-face stimuli. Cereb. Cortex 9, 415430 (1999). subserve attention in macaque areas V2 and V4. J. Neurosci. 19, 17361753
16. Wilson, H. R., Wilkinson, F., Lin, L. M. & Castillo, M. Perception of head (1999).
orientation. Vision Res. (in press). 45. Zipser, K., Lamme, V. A. F. & Schiller, P. H. Contextual modulation in
17. OCraven, K. M., Downing, P. E. & Kanwisher, N. fMRI evidence for objects as primary visual cortex. J. Neurosci. 16, 73767389 (1996).
the units of attentional selection. Nature 401, 584587 (1999). 46. Posner, M. I., Cohen, Y., Choate, L. S., Hockey, R. & Maylor, E. in Preparatory
18. Motter, B. C. Neural correlates of feature selective memory and pop-out in States and Processes (eds. Kornblum, S. & Requin, J.) 4965 (Erlbaum,
extrastriate area V4. J. Neurosci. 14, 21902199 (1994). Hillsdale, New Jersey, 1984).
19. Connor, C. E., Preddie, D. C., Gallant, J. L. & Van Essen, D. C. Spatial attention 47. Maylor, E. A. & Hockey, R. J. Inhibitory component of externally controlled
effects in macaque area V4. J. Neurosci. 17, 32013214 (1997). covert orienting in visual space. J. Exp. Psychol. Hum. Percept. Perform. 11,
20. Luck, S. J., Chelazzi, L., Hillyard, S. A. & Desimone, R. Neural mechanisms of 777787 (1985).
spatial selective attention in areas V1, V2, and V4 of macaque visual cortex. 48. Wilson, H. R. in Spatial Vision (ed. Regan, D.) 6486 (MacMillan, London,
J. Neurophysiol. 77, 2442 (1997). 1991).

176 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Motion perception during saccadic


eye movements
Eric Castet and Guillaume S. Masson

Centre de Recherche en Neurosciences Cognitives (CRNC), UPR 9012 du CNRS31, chemin J. Aiguier13402, Marseille cedex 20, France
Correspondence should be addressed to E.C. (castet@lnf.cnrs-mrs.fr)

During rapid eye movements, motion of the stationary world is generally not perceived despite
displacement of the whole image on the retina. Here we report that during saccades, human
observers sensed visual motion of patterns with low spatial frequency. The effect was greatest when
the stimulus was spatiotemporally optimal for motion detection by the magnocellular pathway.
Adaptation experiments demonstrated dependence of this intrasaccadic motion percept on
2000 Nature America Inc. http://neurosci.nature.com

activation of direction-selective mechanisms. Even two-dimensional complex motion percepts


requiring spatial integration of early motion signals were observed during saccades. These results
indicate that the magnocellular pathway functions during saccades, and that only spatiotemporal
limitations of visual motion perception are important in suppressing awareness of intrasaccadic
motion signals.

During saccadic eye movements, we do not perceive the world The ability to detect a two-frame displacement briefly pre-
as moving. This observation is paradoxical because high-speed sented during the saccade is well studied14,15. It is unclear if dis-
retinal motion can be detected with static eyes, provided it is pro- placement perception involves activation of low-level motion
duced by patterns with low spatial frequency1. Therefore, it is detectors within the magnocellular pathway or a different process,
widely assumed that motion perception is switched off during like attentional tracking of a moving objects successive posi-
saccades. Based on psychophysical results reporting decreased tions16,17. Attempts to assess intrasaccadic motion perception
intrasaccadic sensitivity (for instance, to contrast), two radically using stimuli more specifically linked to early cortical motion
different theories account for the absence of conscious motion processing are scarce. Intrasaccadic retinal stimuli used in most
perception during saccades. The first approach postulates that studies do not optimally activate magnocellular motion detec-
extraretinal signals triggered by saccades actively suppress tors. As a result, human capacity for intrasaccadic motion pro-
intrasaccadic motion processing2,3. Alternatively, some researchers cessing is probably largely underestimated. In brief, visual
propose that visual factors alone might be involved. For exam- functions that might be carried out by the magnocellular stream
ple, intrasaccadic information can be effectively masked by pre- during saccades, including low-level motion processing, are cur-
and postsaccadic visual activations4,5. rently poorly understood.
The general issue of intrasaccadic motion processing remains We present direct evidence for intrasaccadic motion percep-
unclear. Notably, proposals concerning the physiological imple- tion mediated by low-level motion signals transmitted through
mentation of extraretinal suppression raise a number of prob- the magnocellular pathway. Our experiments pinpoint the rele-
lems. The visual system comprises two relatively independent vant visual factors that must be optimized to produce a clear,
pathways from the retina to the cortex. One pathway, which conscious sensation of visual motion during saccades.
extends from the magnocellular subdivision in the lateral genic-
ulate nucleus (LGN) to the parietal cortex, is thought to be RESULTS
important for assessing motion6,7. This magnocellular path- Motion detection
way contains neurons responding optimally to gratings of low The basic stimulus consisted of a vertical grating with low spatial
spatial and high temporal frequencies8,9. It is proposed, based frequency (0.17 cycles per degree) moving at a constant high speed
on psychophysical results, that the magnocellular pathway is (360 per s). With static eyes, the low-contrast grating (10%) was
inactivated during the saccade by selective suppression in the above the critical fusion frequency and was therefore invisible18.
LGN 10,11. However, no physiological evidence supports this While the grating was continuously drifting, observers were
claim. Moreover, neurons in the middle temporal area (MT) of required to make horizontal saccades between two fixation points
awake monkeys can be transiently activated by the visual flow whose spatial separation was varied across trials to alter eye veloc-
created by fixational saccades12, suggesting that precortical sup- ity. The eye speed was concurrently measured in three observers
pression of the motion pathway is unlikely. Furthermore, such who had to report whether or not grating motion was perceived
suppression would be surprising, as there is reason to believe during the saccade (yes/no task). Whenever saccades of moder-
that the essential functions of the magnocellular stream are still ate amplitudes (6) were made in the direction of the grating, a
needed during saccades, whether or not they contribute to con- compelling perception of bars drifting in the direction of the grat-
scious awareness13. Therefore, it was important to investigate ing occurred during the saccade. Mean velocity profiles of sac-
how much intrasaccadic information is available in the mag- cades of different amplitudes for one observer were plotted
nocellular stream. (Fig. 1a). The highest speed reached during a saccade increased

nature neuroscience volume 3 no 2 february 2000 177


2000 Nature America Inc. http://neurosci.nature.com

articles

b
a b
second)

per second)
(/s)
per

(degrees(/s)
velocity
velocity (degrees

Peak velocity
Horizontal

Peak velocity
Horizontal

Saccade
Saccade amplitude
amplitude ()
(degrees)
Time (ms)
Time (ms)

cc
2000 Nature America Inc. http://neurosci.nature.com

of perceiving
perceiving motion
Probability of
motion
Probability

Peak velocity
Peak velocity (/s)
(degrees per second)

Fig. 1. Motion perception during a saccade in the same direction as a high-speed vertical grating (10% contrast). (a) Eye speed during leftward sac-
cades (observer EC) as a function of time and saccade amplitude. Dashed line shows constant speed of a leftward-moving grating. For each saccade
amplitude (two indicated by arrows), the mean velocity profile was obtained from aligning onsets of individual saccades and averaging their velocity
profiles. (b) Dependence of peak velocity on saccade amplitude. For each combination of saccade amplitude and grating speed (solid symbols, 360
per s; open symbols, 300 per s), we measured the mean saccadic peak velocity by averaging individual peak velocities. (c) Probability of perceiving
motion as a function of saccadic peak velocity for three observers. Gratings moving at either 360 per s (filled symbols) or 300 per s (open symbols)
were presented in successive blocks. Error bars to right show average s.e. For all observers, the two inverted U-shaped curves were laterally shifted
by 60 per s (horizontal arrows).

with saccade amplitude19,20 (Fig. 1b). The probability of perceiv- partly explains the usual impairment of intrasaccadic motion
ing motion depended on this saccadic peak velocity (Fig. 1c). With processing. This derives from two fundamental spatiotemporal
gratings either at 360 per s (solid symbols) or 300 per s (open characteristics of motion detection. First, individual low-level
symbols), the data curves resembled an inverted U and were lat- motion detectors only respond to a restricted range of spa-
erally shifted by a peak-velocity difference of about 60 per s (hor- tiotemporal frequencies2123, and second, they need a minimum
izontal arrows). This value corresponded to the difference between amount of time to integrate motion energy24. Thus, for any spa-
the two grating speeds. These results strongly suggest that motion tial-frequency channel, the temporal frequency at the retinal
perception depends on the difference between the peak eye veloc- level changes too rapidly to provide a reliable signal, at least dur-
ity and grating speed; the relevant factor is the speed (or tempo- ing the early acceleration and late deceleration phases of the
ral frequency) of the grating relative to the retina. This is best seen velocity profile. However, within a brief period during which
when data from Fig. 1c are replotted as a function of the retinal the velocity peaks, retinal temporal frequency is relatively con-
temporal frequency at the peak of the saccade (Fig. 2a). Here the stant. It is during this critical period that motion detectors could
two curves are superimposed, peaking between 10 and 25 Hz, be effectively activated, provided the spatiotemporal combina-
depending on the observer. tion were optimal. This hypothesis is fully consistent with our
The phenomenon can be understood if we look at the effec- motion effect. With small saccades, the temporal frequency of
tive spatiotemporal stimulus at the retinal level. The retinal speed the grating relative to the retina was too high even when the peak
(and temporal frequency) of the grating was high at the begin- velocity was reached (Fig. 2a). For instance, with a 2 saccade,
ning of the saccade and decreased until the peak was reached. the retinal temporal frequency at the peak was about 40 Hz (240
After the peak, retinal speed increased again (Fig. 1a). We pro- per s), much too close to the fusion frequency to produce a clear
pose that this constantly changing retinal temporal frequency motion percept. With medium saccades, motion perception was

178 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

optimal, because these sac- a


cades yielded retinal tempo-

perceivingmotion
motion
ral frequencies around the

of
Probability of
peak that fell within the opti-

Probability
mal temporal range for

perceiving
motion detection 25 . With
large saccades (when the
peak velocity approaches the
grating speednull relative
speed), static visual channels
are optimally activated. Peak retinal temporal frequency (Hz)
Observers report that the
sense of motion is lost and
replaced by the perception of b
Fig. 2. Temporal tuning of motion perception. (a) Data in Fig.
a flashed grating. A previ-
1c replotted as a function of retinal temporal frequency at the
Response (impulses/sec)

ous study has shown that


(impulses/s)

peak of the saccade (circles). Positive and negative abscissa val-


stabilizing the retinal image ues denote retinal motion in the same direction as the saccade
of a rapidly moving grating
2000 Nature America Inc. http://neurosci.nature.com

and in the opposite direction, respectively. Triangles, data


with a saccade produces a obtained for observer EC during fixation. The stimulus was a
static flash percept26. How- 25-ms movie (4 frames) crudely approximating the different
Response

ever, observers did not retinal temporal frequencies experienced around the peak
report motion perception, velocity of the saccade. The grating contrast was either 10%
presumably because the sin- (solid triangles) or 5% (open triangles). (b) Temporal tuning
gle spatial frequency used curve of a V1 magnocellular cell measured with a 0.9-cycle-per-
degree grating (replotted from ref. 8). This cell is direction
(1 cycle per degree) was too selective and projects to area MT.
Temporal frequency (Hz)
high. Beside making stimuli
non-optimal for the magno-
cellular pathway, high spatial
frequencies present another
problem: during a saccade, the range of retinal temporal fre- We showed that a saccade toward a high-speed grating of low
quencies swept through over a given period increases propor- spatial frequency could produce a clear motion percept: the grat-
tionally with spatial frequency. As a result, the restricted range of ing briefly appeared to move in the direction of the saccade. This
temporal frequencies required for motion processing within a percept was optimal when the retinal stimulus was within the range
single temporal-frequency channel is encountered for shorter for motion detection during the period including peak velocity.
periods as spatial frequency is increased. Thus, this period is The optimal retinal temporal frequencies for motion detection
approximately sixfold shorter in the previous study26 than in (1025 Hz; Fig. 2b) were consistent with the broad and high-cutoff
ours, possibly accounting for the failure to perceive intrasac- tuning curves of direction-selective cells in primate striate cortex8,9.
cadic motion with high spatial frequencies.
Processing intrasaccadic visual motion under our protocol Direction discrimination
requires integrating different speeds across time, presumably Possible inconsistency in criteria for motion percepts across
by some form of averaging, within a brief period of 25 ms. observers and runs may be a weakness of the previous study. This
We tested the ability to process this kind of visual stimulus with may explain why peak values of the inverted U-curves differed
static eyes. For each saccade amplitude previously tested, a 25- for different observers (Fig. 2a). To overcome this, we also mea-
ms stimulus with 3 different grating speeds was created to sured thresholds for direction discrimination. Because direction
approximate the variation in speeds experienced near the sac- selectivity is an essential property of early motion detectors, a
cadic peak velocity (see Methods). Again, subjects indicated direction discrimination task further probed the involvement of
whether or not motion was perceived. The results were quali- motion pathways in intrasaccadic motion perception.
tatively similar to that obtained during saccades: for low con- We predicted that perceived direction would oppose the direc-
trasts, probability of perceiving motion as a function of peak tion of the saccade when the peak of the velocity profile exceed-
retinal temporal frequency was an inverted U-shaped curve. ed the grating speed for a sufficiently long duration. To test this,
Quantitative differences were demonstrated by observer EC a grating whose speed (or equivalent temporal frequency) was
(triangles, Fig. 2a). The leftward shift of the curve probably controlled by an adaptive-staircase procedure was presented on
reflects the different perceptual criterion used for this task. each trial as before. Observers made large saccades of constant
Contrast sensitivity was also higher in the static eye condi- amplitude (14) and reported whether perceived motion was in
tion: with a 10% contrast grating (as in the intrasaccadic task), the same direction as the saccade (forward response) or its oppo-
high temporal frequencies were not above fusion frequency site (backward response). Using 2 staircases, we could assess the
and, therefore, produced motion perception (solid triangles). grating speeds producing forward responses with probabilities
By halving contrast, motion perception was decreased for these of 0.29 and 0.71. Backward percepts appeared less vivid than for-
high frequencies, yielding an inverted U-curve (open trian- ward percepts, presumably because of concurrent activation of
gles). It is impossible to determine whether this sensitivity dif- static channels during the two brief periods when eye and grating
ference results from extraretinal or visual factors, mainly speeds matched. The point of subjective stationarity (PSS) was
because the effective retinal stimulus differs between experi- determined by tracking the speed of a grating that produced a
mental conditions. static percept (equiprobable forward and backward responses).

nature neuroscience volume 3 no 2 february 2000 179


2000 Nature America Inc. http://neurosci.nature.com

articles

Threshold (degrees per second)

PSS = 355/s PSS = 358/s PSS = 347/s


Direction-specific adaptation
Although the spatiotemporal selectivity of
these motion effects suggests the involvement
of early motion detectors, one could argue
that the percept is merely due to static mech-
anisms recruited to detect a temporal change
of position16,17. To rule out this possibility, we
used a classic protocol for direction-specific
adaptation27. A vertical high-contrast adapt-
Forward

Backward

Forward

Backward

Forward

Backward
ing grating (12 Hz, 0.17 cycles per degree, 72
Forward

Forward
Forward
Backward

Backward
Backward
per s) covering the whole screen was present-
ed while observers stared at a fixation point.
Its temporal frequency was chosen to opti-
mally activate motion detectors25. During the
Fig. 3. Direction discrimination experiment. Thresholds for perceiving motion either in the
direction of the saccade (forward) or in the opposite direction (backward). Forward and back-
test period, a vertical grating (50 Hz, 0.17
ward thresholds respectively correspond to grating speeds larger and smaller than the average cycles per degree, 300 per s) of variable con-
saccadic peak velocity. trast presented in the top or bottom half of
the screen was moved either in or opposite to
2000 Nature America Inc. http://neurosci.nature.com

the adapting direction. Observers were


required to make saccades in the same direc-
Thresholds for all observers lay within a 2535 per s range tion as the test grating and to indicate the part of the screen in
(46 Hz): forward differential thresholds correspond to the dif- which the intrasaccadic motion was perceived. The saccade
ference between 0.71 and 0.50 levels, and backward thresholds amplitude for each observer was chosen to optimize motion
correspond to the 0.500.29 difference (Fig. 3). perception (as assessed in the first experiment). Contrast
For comparison, we also measured these direction-discrimi- thresholds were dramatically increased when the adapting and
nation thresholds during fixation. The stimulus was a 25-ms movie test gratings were in the same direction, whereas adaptation in
of 4 frames simulating 3 successive speeds around the peak of a the opposite direction had only a slightly detrimental effect
14 saccade. We varied the average speed from a few degrees per (Fig. 4; comparison of across-observer means, t 2 = 8;
second to the left to a few degrees per second to the right. Mea- p = 0.015). This direction-specific threshold elevation is a clas-
sured thresholds (observers E.C., G.S.M., data not shown) were sic signature of an early direction-sensitive mechanism27. In
four- to sixfold lower than those during saccades (Fig. 3). Howev- primates, direction selectivity is a visual property that starts at
er, it is impossible to assess whether this decrease was related to the level of the primary visual cortex28. Therefore, this finding
extraretinal signals or to visual factors alone, especially as retinal is at odds with suggestions that the activity of the magnocel-
stimuli differed between conditions. One clear difference was that, lular pathway (constituting the dominant input to motion
because of the large saccade, the effective stimulus width for the detectors 6,7) is suppressed at a precortical level during sac-
intrasaccadic condition was narrower (about 12) than the full cades10,11.
screen available during the fixation condition (27). As a result,
spatial integration mechanisms could not be as efficient as during Two-dimensional motion perception
fixation. Moreover, the screen edges sweeping over peripheral reti- In the experiments above, the perceived direction of the
na may produce lateral masking. Intrasaccadic thresholds may also intrasaccadic percept always matched the axis of the saccade
be inflated by variability in peak saccade speed across trials, as we itself. This coincidence raises the possibility that the percept
used average, rather than trial-by-trial, eye speed. Therefore, inves- was actually determined by an extraretinal signal associated
tigating the relative influence of visual versus
extraretinal factors on direction discrimina-
tion during saccades requires further research.
threshold

Here, the relevant difference between fix-


Contrast threshold

ation and intrasaccadic thresholds concerned


elevation

the PSS. During saccades, the PSS (above each


elevation

graph in Fig. 3) was smaller than the average


Contrast

saccadic peak velocity (370 per s), so the cor-


responding retinal speed averaged over 25 ms
was close to zero (mean, 6 per s). This sug-
gests that stationarity is perceived when reti-
nal speeds within 2530 ms are of opposite
directions and thus approximately average to
zero, consistent with our measurement of the Opposite
PSS during fixation at nearly 0 per s. Same
In summary, the intrasaccadic ability to
Fig. 4. Direction-selective adaptation. Observers made horizontal saccades and detected
discriminate between forward and back- whether a horizontally moving test grating was located in the top or bottom half of the screen
ward directions supports our hypothesis that (spatial 2AFC). Contrast sensitivity was measured with or without adaptation to a moving grat-
direction signals relevant to the task are ing. The data clearly show direction-selective adaptation: contrast-threshold elevation (relative
indeed extracted during a short period in to the unadapted condition) was larger when the adapting grating was in the same rather than in
which eye velocity peaks. the opposite direction.

180 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a b
2000 Nature America Inc. http://neurosci.nature.com

Fig. 5. Perceived direction of two-dimensional motion. (a) Stimuli were oblique (45) moving gratings (contrast 30%; temporal frequency as before)
presented within square (20 20) or rectangular (20 10) apertures. As before, observers made horizontal saccades of optimal amplitude and
then rotated an arrow in the middle of the screen to indicate the direction of perceived intrasaccadic motion. (b) Distributions of perceived direc-
tions in polar coordinates. The origin (0) of the angular axis represents the motion component perpendicular to the bars of the grating. The radial
axis plots, in log coordinates, the number of observations collected for each direction. Mean direction is given by . With a square aperture, per-
ceived directions cluster around the perpendicular component. With a rectangular aperture, perceived directions are parallel to the long axis (barber-
pole illusion).

with the saccade14. To avoid this confounding effect, we used DISCUSSION


the display from the first experiment but rotated the moving We show that making saccades in the direction of a rapidly mov-
grating by 45 and presented it within a large square aperture ing grating produced clear motion percepts when retinal image
(Fig. 5a, top). Observers made horizontal saccades of optimal motion (averaged over a critical period around the saccadic peak
amplitudes, and then adjusted the direction of an arrow pre- velocity) was within the motion-detection range. Observers could
sented on the screen to report the perceived direction of the distinguish between retinal motion in the direction of the sac-
intrasaccadic percept. The distribution of the perceived direc- cade and retinal motion in the opposite direction, and perceived
tions was centered around the grating direction (Fig. 5b, top), a static grating when retinal speed averaged over 2530 ms was
as would be expected if motion detectors with the preferred close to null. Adapting during fixation to a low spatial and high
oblique orientation matching the grating orientation were acti- temporal frequency grating produced a direction-specific reduc-
vated in this condition. tion in contrast sensitivity to intrasaccadic motion perception.
Furthermore, motion perception of real complex objects Additionally, spatial integration of early motion signals leading to
requires the spatial integration of different local motion sig- two-dimensional motion perception was still observed during
nals that are not in the direction of the eye movements29,30. If saccades. We conclude that these intrasaccadic motion percepts
early motion signals are not suppressed during saccades, as revealed the activation of direction-selective mechanisms tuned
suggested by previous experiments, one could argue instead to low spatial and high temporal frequencies within the magno-
that integration of these signals into a two-dimensional veloc- cellular pathway.
ity signal is actively suppressed. A classic stimulus used to study We designed the stimulus to optimize intrasaccadic motion
motion integration is the barber-pole illusion 31 : when an perception. First, as the grating was invisible at the beginning
oblique grating moves within a rectangular, rather than square, (and at the end) of each saccade, there was no pre- or post-sac-
aperture, its perceived motion is parallel to the long side of the cadic visual information that might have produced forward or
aperture. This phenomenon, which indicates the importance backward masking4,5,36,37. Moreover, in contrast to studies using
of the motion signals elicited near aperture borders32,33, was a two-frame sequence, the grating was not abruptly presented
also tested. The height of the aperture used before was simply during the saccade. Instead, it was continuously displayed on
halved to obtain a horizontal barber-pole (Fig. 5a, bottom). each trial. This point was crucial: the gradual transitions between
All observers perceived the classic barber-pole illusionhere, a retinal temporal range above fusion frequency and an optimal
in a horizontal direction (Fig. 5b, bottom). Intrasaccadic range for motion detection minimized the temporal energy
motion perception based on activation of low-level detectors spread and the intrusion of masking transient signals. This opti-
tuned to all possible directions followed by integration of the mization allowed a study of motion detection per se rather than
resulting signals, presumably at a second stage29,34,35, is therefore displacement detection. The involvement of early motion pro-
possible during saccades. cessing is unclear in numerous studies of intrasaccadic displace-

nature neuroscience volume 3 no 2 february 2000 181


2000 Nature America Inc. http://neurosci.nature.com

articles

ment perception14,15. Two-frame presentations used in displace- METHODS


ment studies are weak stimuli for motion processing. As a con- Psychophysics. Stimuli were displayed on a 21-inch Sony color monitor
sequence, it is possible that performance in these studies might (GDM-4011P) driven by a display controller (Cambridge Research Sys-
rely exclusively on visual processes that detect changes in posi- tem VSG 2/3F, Rochester, UK) with a 160-Hz frame rate. At a viewing
tion over time16,17. distance of 68 cm, the average horizontal separation between adjacent
pixels subtended 0.035 of visual angle. The screen subtended 27 21.
In our experiments, intrasaccadic motion perception was A lookup table in the software was used to linearize the intensity response
easily experienced when magnocellular motion detection was of the screen phosphors at an 8-bit luminance resolution.
optimized. In primates, the first stage of motion processing
takes place in the striate cortex (V1). The one-dimensional Stimuli displayed during saccades. On each trial, a moving grating of
edge-motion signals extracted in V1 are thought to be inte- low spatial frequency (0.17 cycles per degree) was displayed for 2 s. Dur-
grated at a later stage into a two-dimensional velocity signal ing the first 500 ms, the contrast increased using a raised cosine func-
within the middle temporal area34,35. There is ample evidence tion from 0% to the desired level, which was kept constant for 1000 ms.
that motion perception depends on the activity of MT cells3840. The contrast then decayed to 0% using the same function during the last
In addition, MT cells of alert monkeys consistently respond to 500 ms. A low-pitch sound indicated that the contrast had reached the
directional signals induced by randomly directed small-ampli- desired level, prompting saccades. A high-pitch sound, presented when
the grating disappeared, prompted observers to respond. The temporal
tude (about 0.8) saccades while monkeys fixate12. MT neurons
frequency was either 50 Hz (300 per s) or 60 Hz (360 per s) except in the
transiently increase their firing rates when these fixational sac- second experiment (direction discrimination), where it was varied with
cades induce a retinal flow in the neurons preferred direction,
2000 Nature America Inc. http://neurosci.nature.com

an adaptive procedure. The mean luminance of the grating was 22 cd per


whereas neuronal responses are mildly suppressed when sac- m2 (4.5 cd per m2 in the second experiment). Observers AL and YR were
cades induce null retinal flow. In this framework, the spa- naive as to the purpose of the experiments.
tiotemporal characteristics of the effects in experiments 13
suggest the involvement of direction-selective V1 neurons8,9, Stimuli displayed with static eyes. A 25-ms movie was presented at 3 dif-
whereas motion integration observed in experiment 4 was prob- ferent speeds during fixation by displaying 4 frames of a grating whose
ably related to MT responses. spatial phase was appropriately shifted. For each saccade amplitude (test-
Our findings suggest that visual factors are the essential ed in the first experiment), 3 retinal speeds centered on the saccadic peak
determinants of suppression of consciousness of motion dur- velocity were calculated from the saccadic velocity profiles over 25 ms.
For instance, for a 2 amplitude, the 3 successive retinal speeds of the
ing saccades4,5,36,37. Clearly, extraretinal signals do not switch grating were S1, 270 per s (6 ms before the peak), S2, 240 per s (at the
off motion processing. Studies suggesting such suppression peak) and S1 again (6 ms after the peak); average speed, 260 per s.
actually measured the visibility of static gratings (0 Hz)10,11, and
therefore are not inconsistent with our study, which used spa- Measurement of eye movements. Horizontal movements of the right eye
tiotemporal stimuli optimized for motion detection by the mag- were recorded using a high-resolution infrared scleral-reflectance system
nocellular pathway (with low spatial and high temporal (IRIS Skalar, Delft, Netherlands). Analog signals were digitized at 500
frequencies). Hz with a 12-bit resolution ADC. A bite bar was used to restrain head
Our results also pertain to the issue of perceptual stability movements. Two points of different colors indicated the size and the
during eye movements in relation to extraretinal signals14,15. In direction of the saccade to be made. On each trial, a sound instructed
our experiments, it was striking that a static flash percept observers to make the saccade. Data were analyzed off line. Inappropri-
ate saccades were discarded from the analysis.
occurred when the eye peak velocity equaled the speed of the
moving grating. Such a static appearance was inconsistent with
Direction discrimination. Across trials of the same block, the direction of
the idea that extraretinal signals are used to compensate for the the saccade (14 amplitude) was constant (leftward or rightward).
effect of eye movements, as generally assumed with smooth eye Observers were required to indicate whether perceived motion of the
movements 4144. This hypothesis would predict high-speed grating was in the same direction as the saccade (forward) or in the oppo-
motion perception of the grating. Therefore, it seems that sac- site direction (backward). The grating (20% contrast) covered the whole
cadic eye movements invoke other mechanisms to achieve per- screen and had its temporal frequency controlled by one of 3 staircases
ceptual stability 13,45 . Alternatively, it is possible that a with steps of 0.5 Hz50. One staircase (starting value, 360 per s; 60 Hz)
recalibration process due to extraretinal signals acts more slow- converged on a 0.50 threshold indicating the speed at which the grating
ly than the actual saccadic eye movements46, or that extraretinal appeared static. The two other staircases tracked the 0.29 and 0.71 thresh-
signals carry a damped representation of eye position47. olds. Their starting values, which were 300 per s (50 Hz) and 420 per s
(70 Hz) in the training period, were respectively increased and lowered as
Here we showed that the magnocellular pathway functions performance improved across blocks. The actual starting values were
during saccades. Indeed, suppression of the magnocellular- randomly chosen within a range of 2 Hz centered on the above values.
dominated stream would have important adverse effects48. For The end of a block occurred when a minimum of nine reversals had been
instance, the role of the parietal cortex in visual attention49 completed in each staircase. The three thresholds were then estimated
makes it well suited to alert and orient the visual system toward by averaging the six last reversals in each staircase. The same direction
a peripheral transient signal detected during the saccade. Given differential threshold (DT) was defined as the difference between the
the high frequency of saccades in normal vision, future research 0.71 and 0.50 levels, and the opposite direction DT as the difference
should examine the behavioral importance of processing between the 0.50 and 0.29 levels.
intrasaccadic visual information.
Our findings do not rule out the possibility that an extrareti- Direction-specific adaptation. Required saccade direction (leftward or
rightward, in the direction of the test grating moving at 300 per s) was
nal signal alters intrasaccadic visual motion processing. Howev-
randomly alternated across trials. For each observer, a constant saccade
er, we clearly showed that such a putative extraretinal signal does amplitude was chosen from results of the first experiment to optimize
not suppress motion perception during saccades. Therefore, it is motion perception (4 for EC and 3 for AL and YR). The contrast of the
more parsimonious to interpret the usual absence of intrasac- test grating was adjusted with an adaptive staircase procedure converging
cadic motion perception as the result of visual and/or attention- on a 0.71 threshold (steps of 0.4% contrast)50. Two staircases, each
al factors alone. assigned to a grating direction, were randomly interleaved within a block.

182 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Contrast thresholds were measured with or without adaptation to a mov- 17. Cavanagh, P. Attention-based motion perception. Science 257, 15631565
ing grating (80% contrast), which covered the whole screen and had the (1992).
18. Robson, J. G. Spatial and temporal contrast sensitivity functions of the visual
same spatial frequency as the test grating and a temporal frequency of system. J. Opt. Soc. Am. 56, 11411142 (1966).
12 Hz (optimal for motion detection of low spatial frequencies25). The 19. Bahill, A. T., Clark, M. R. & Stark, L. The main sequence, a tool for studying
adapting grating, whose direction was constant within any block, was human eye movements. Math. Biosci. 24, 191204 (1975).
presented for one minute at the beginning of each block. Observers were 20. Collewijn, H., Erkelens, C. J. & Steinman, R. M. Binocular co-ordination of
then exposed to cycles of test periods (during which one saccade had to human horizontal saccadic eye movements. J. Physiol. (Lond.) 404, 157182
(1988).
be made within 1500 ms) and 10-s adapting periods. Test gratings were 21. Morgan, M. J. Analogue models of motion perception. Phil. Trans. R. Soc.
presented either in the upper or lower part of the screen (spatial 2AFC). Lond. B Biol. Sci. 290, 117135 (1980).
A block ended when a minimum of eight reversals had been completed in 22. van Santen, J. P. & Sperling, G. Elaborated Reichardt detectors. J. Opt. Soc.
each staircase. The two thresholds were then estimated by averaging the Am. A Opt. Image Sci. Vis. 2, 300321 (1985).
23. Watson, A. B. & Ahumada, A. J., Jr. Model of human visual-motion sensing.
last six reversals in each staircase. J. Opt. Soc. Am. A Opt. Image Sci. Vis. 2, 322341 (1985).
24. Burr, D. C. Temporal summation of moving images by the human visual
Two-dimensional motion perception. For a given saccade direction (left- system. Proc. R. Soc. Lond. B Biol. Sci. 211, 321339 (1981).
ward or rightward), the orientation of the grating was randomized so 25. Anderson, S. J. & Burr, D. C. Spatial and temporal selectivity of the human
motion detection system. Vision Res. 25, 11471154 (1985).
that the motion component perpendicular to the bars was either above or 26. Deubel, H., Elsner, T. & Hauske, G. Saccadic eye movements and the
below horizontal. The two different saccade directions were presented detection of fast-moving gratings. Biol. Cybern. 57, 3745 (1987).
in distinct blocks. The perceived directions plotted in Fig. 5b were aver- 27. Levinson, E. & Sekuler, R. The independence of channels in human vision
aged over the four possible quadrants (two for each saccade direction). selective for direction of movement. J. Physiol. (Lond.) 250, 347366 (1975).
The square and rectangular aperture conditions were randomly inter- 28. Hubel, D. H. & Wiesel, T. N. Functional architecture of macaque monkey
2000 Nature America Inc. http://neurosci.nature.com

visual cortex. Proc. R. Soc. Lond. B Biol. Sci. 198, 159 (1977).
leaved within each block. 29. Adelson, E. H. & Movshon, J. A. Phenomenal coherence of moving visual
patterns. Nature 300, 523525 (1982).
30. Yuille, A. L. & Grzywacz, N. M. A computational theory for the perception of
ACKNOWLEDGEMENTS coherent visual motion. Nature 333, 7174 (1988).
We thank L. S. Stone, M. J. Morgan, J. K. ORegan and A. Riehle for comments 31. Wuerger, S., Shapley, R. & Rubin, N. On the visually perceived direction of
on the manuscript. motion by Hans Wallach: 60 years later. Perception 11, 13171367 (1996).
32. Kooi, F. L. Local direction of edge motion causes and abolishes the barber
pole illusion. Vision Res. 33, 23472351 (1993).
RECEIVED 19 AUGUST; ACCEPTED 8 DECEMBER 1999 33. Castet, E., Charton, V. & Dufour, A. The extrinsic/intrinsic classification of
2D motion signals with barber-pole stimuli. Vision Res. 39, 915932 (1999).
34. Albright, T. D. & Stoner, G. R. Visual motion perception. Proc. Natl. Acad. Sci. USA
1. Burr, D. C. & Ross, J. Contrast sensitivity at high velocities. Vision Res. 22, 92, 24332440 (1995).
479484 (1982). 35. Movshon, A. J., Adelson, E. H., Gizzi, M. S. & Newsome, W. T. in Pattern
2. Shioiri, S. & Cavanagh, P. Saccadic suppression of low-level motion. Vision Recognition Mechanisms (eds. Chagas, C., Gattas, R. & Gross, C. G.) 117151
Res. 29, 915928 (1989). (Vatican Press, Rome, 1985).
3. Ilg, U. J. & Hoffmann, K. P. Motion perception during saccades. Vision Res. 36. Breitmeyer, B. G. & Ganz, L. Implications of sustained and transient channels
33, 211220 (1993). for theories of visual pattern masking, saccadic suppression, and information
4. Judge, S. J., Wurtz, R. H. & Richmond, B. J. Vision during saccadic eye processing. Psychol. Rev. 83, 136 (1976).
movements. I. Visual interactions in striate cortex. J. Neurophysiol. 43, 37. Volkmann, F. C. Human visual suppression. Vision Res. 26, 14011416
11331155 (1980). (1986).
5. Campbell, F. W. & Wurtz, R. H. Saccadic omission: why we do not see a grey- 38. Salzman, C. D., Britten, K. H. & Newsome, W. T. Cortical microstimulation
out during a saccadic eye movement. Vision Res. 18, 12971303 (1978). influences perceptual judgements of motion direction. Nature 346, 174177
6. Maunsell, J. H., Nealey, T. A. & DePriest, D. D. Magnocellular and (1990).
parvocellular contributions to responses in the middle temporal visual area 39. Britten, K. H., Newsome, W. T., Shadlen, M. N., Celebrini, S. & Movshon,
(MT) of the macaque monkey. J. Neurosci. 10, 33233334 (1990). J. A. A relationship between behavioral choice and the visual responses of
7. Merigan, W. H., Byrne, C. E. & Maunsell, J. H. Does primate motion neurons in macaque MT. Vis. Neurosci. 13, 87100 (1996).
perception depend on the magnocellular pathway? J. Neurosci. 11, 34223429 40. Pasternak, T. & Merigan, W. H. Motion perception following lesions of the
(1991). superior temporal sulcus in the monkey. Cereb. Cortex 4, 247259 (1994).
8. Movshon, J. A. & Newsome, W. T. Visual response properties of striate 41. Newsome, W. T. & Wurtz, R. H. Probing visual cortical function with discrete
cortical neurons projecting to area MT in macaque monkeys. J. Neurosci. 16, chemical lesions. Trends Neurosci. 11, 394400 (1988).
77337741 (1996). 42. Erickson, R. G. & Thier, P. A neuronal correlate of spatial stability during
9. Hawken, M. J., Shapley, R. M. & Grosof, D. H. Temporal-frequency selectivity periods of self-induced visual motion. Exp. Brain Res. 86, 608616 (1991).
in monkey visual cortex. Vis. Neurosci. 13, 477492 (1996). 43. Haarmeier, T., Thier, P., Repnow, M. & Petersen, D. False perception of
10. Ross, J., Burr, D. & Morrone, C. Suppression of the magnocellular pathway motion in a patient who cannot compensate for eye movements. Nature 389,
during saccades. Behav. Brain Res. 80, 18 (1996). 849852 (1997).
11. Burr, D. C., Morrone, M. C. & Ross, J. Selective suppression of the 44. Matin, L. et al. Oculoparalytic illusion: visual-field dependent spatial
magnocellular visual pathway during saccadic eye movements. Nature 371, mislocalizations by humans partially paralyzed with curare. Science 216,
511513 (1994). 198201 (1982).
12. Bair, W. & OKeefe, L. P. The influence of fixational eye movements on the 45. Deubel, H., Bridgeman, B. & Schneider, W. X. Immediate post-saccadic
response of neurons in area MT of the macaque. Vis. Neurosci. 15, 779786 information mediates space constancy. Vision Res. 38, 31473159 (1998).
(1998). 46. Grsser, O. J., Krizic, A. & Weiss, L. R. Afterimage movement during saccades
13. ORegan, J. K. Solving the real mysteries of visual perception: the world as an in the dark. Vision Res. 27, 215226 (1987).
outside memory. Can. J. Psychol. 46, 461488 (1992). 47. Bockisch, C. J. & Miller, J. M. Different motor systems use similar damped
14. Bridgeman, B., Van der Heijden, A. H. C. & Velichkovsky, B. M. A theory of extraretinal eye position information. Vision Res. 39, 10251038 (1999).
visual stability across saccadic eye movements. Behav. Brain Sci. 17, 247292 48. Van Essen, D. C. & DeYoe, E. A. in The Cognitive Neurosciences (ed.
(1994). Gazzaniga, M. S.) 383400 (MIT Press, Cambridge, Massachusetts, 1995).
15. Li, W. & Matin, L. Saccadic suppression of displacement: separate influences of 49. Corbetta, M., Miezin, F. M., Shulman, G. L. & Petersen, S. E. A PET study of
saccade size and of target retinal eccentricity. Vision Res. 37, 17791797 (1997). visuospatial attention. J. Neurosci. 13, 12021226 (1993).
16. Braddick, O. J. Low-level and high-level processes in apparent motion. 50. Levitt, H. Transformed up-down methods in psychoacoustics. J. Acoust. Soc. Am.
Phil. Trans. R. Soc. Lond. B Biol.Sci. 290, 137151 (1980). 49, 467477 (1971).

nature neuroscience volume 3 no 2 february 2000 183


2000 Nature America Inc. http://neurosci.nature.com

articles

Thermosensory activation of insular


cortex
A. D. Craig1, K. Chen2,3, D. Bandy2 and E. M. Reiman2,4

1 Division of Neurosurgery, Barrow Neurological Institute, 350 West Thomas Rd., Phoenix, Arizona 85013, USA
2 Positron Emission Tomography Center, Good Samaritan Regional Medical Center, Phoenix, Arizona 85006, USA
3 Department of Mathematics, Arizona State University, Tempe, Arizona 85280, USA
4 Department of Psychiatry, University of Arizona, Tucson, Arizona 85721, USA
Correspondence should be addressed to A.D.C. (bcraig@mha.chw.edu)

Temperature sensation is regarded as a submodality of touch, but evidence suggests involvement of


2000 Nature America Inc. http://neurosci.nature.com

insular cortex rather than parietal somatosensory cortices. Using positron emission tomography
(PET), we found contralateral activity correlated with graded cooling stimuli only in the dorsal
margin of the middle/posterior insula in humans. This corresponds to the thermoreceptive- and
nociceptive-specific lamina I spinothalamocortical pathway in monkeys, and can be considered an
enteroceptive area within limbic sensory cortex. Because lesions at this site can produce the post-
stroke central pain syndrome, this finding supports the proposal that central pain results from loss
of the normal inhibition of pain by cold. Notably, perceived thermal intensity was well correlated
with activation in the right (ipsilateral) anterior insular and orbitofrontal cortices.

The cutaneous sense of temperature is a distinct form of somat- indicates that a dedicated thalamic nucleus relays topographic, dis-
ic sensibility mediated by specific primary afferent receptors, yet criminative thermoreceptive-specific and nociceptive-specific lam-
its representation in the brain is unknown1,2. Thermoreception ina I spino- and trigeminothalamic projections to the dorsal
has two aspects, an exteroceptive, discriminative aspect impor- margin of middle/posterior insular cortex2,12,13. Microstimulation
tant for object recognition and environmental exploration and seemingly localized to the homologous thalamic region in awake
an enteroceptive, affective aspect important for autonomic activ- humans can produce discrete, graded sensations of cold14. Data
ity, homeostasis and thermoregulatory behavior. Whereas the from a PET imaging study of the thermal grill illusion of pain4 and
former is more obvious and traditionally emphasized, the latter an fMRI study14 indicate that strong innocuous cool stimulation
is an essential feature of mammalian physiology. Conceptual (20C) activates contralateral insular cortex, but not parietal
recognition that thermal sensibility is an emergent aspect of ente- somatosensory cortical areas.
roception (the sense of the physiological condition of the body We used PET imaging of regional cerebral blood flow (rCBF)
itself) would accomodate a variety of considerations and have to examine changes in forebrain activity directly related to the
strong implications for other sensations from the body typically intensity of graded cooling stimuli. Our results demonstrate that
regarded as exteroceptive, for instance, the sensation of pain. discriminative thermosensory cortex lies in the dorsal margin of
Temperature and pain sensations are intimately associated the middle/posterior insula of humans.
functionally and anatomically in the central nervous system
consistent with their common importance for maintenance of RESULTS
body integrityand they are accordingly integrated. A funda- We addressed the hypothesis that thermal stimuli activate the con-
mental interaction, the cold inhibition of pain, is a well estab- tralateral insular cortex by examining rCBF changes with graded
lished therapeutic phenomenon with a demonstrable central cooling of the right hand. Tonic thermal stimuli were used,
basis3,4. Loss of this interaction, which may result in the disinhi- because the tonic responses of thermoreceptive-specific lamina I
bition of pain, is proposed as a possible cause of the post-stroke neurons linearly encode innocuous cool temperatures, whereas
central pain syndrome5. This syndrome is generally character- their dynamic responses saturate more quickly1,15,16. Thus, the
ized by intractable burning or aching pain referred to deep and stimuli ramped slowly downward and achieved a stable target
cutaneous tissues within a region of paradoxical hypalgesia and temperature 15 seconds before scan initiation. Preliminary testing
thermal hypesthesia. (It occurs in 28% of stroke patients and verified the discriminability of these stimuli and the stability of
2540% of spinal cord injury and multiple sclerosis patients; it the evoked sensations over the 4560-second stimulation peri-
is unresponsive to opiates.) Brain lesions that produce this syn- od1. Using a verbal, open-ended zero to ten scale for magnitude
drome interrupt the ascending spinothalamic pathway respon- estimation of cold intensity, all volunteers were able to differentiate
sible for pain and temperature sensations6,7, and the critical stimuli that differed by 2C in ascending or descending sequences,
cortical locus lies in a parieto-insular region8. and their ratings were correlated with stimulus intensity (Fig. 1a).
Other findings directly suggest that thermal sensation is rep- We first explored changes in regional activity over the entire
resented in insular cortex, an area usually associated with auto- forebrain by examining differences (subtractions) between the var-
nomic and limbic function911. Functional anatomy in monkeys ious thermal stimulus conditions (30C, 28C, 26C, 24C, 20C)

184 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

a c

Activity
Ratings

Temperature (C) Temperature (C)

b Fig. 1. Regression plots. (a) Average ratings given by the volunteers for the
thermal stimuli during the PET scans (r = 0.83; p < 106). (b) Average rCBF
2000 Nature America Inc. http://neurosci.nature.com

activity in left (contralateral) insular cortex (coordinates = 36, 22, 24; r =


0.55; p < 0.00005). (c) Step-like shift of average rCBF activity in right poste-
Activity

rior parietal cortex (coordinates = 36, 40, 36). Bars indicate standard error.

sion, but there were obvious shifts in magnitudes and spatial extents.
The largest correlations with ratings were located in the right
orbitofrontal and anterior insular regions (Table 2; Fig. 2). A sig-
nificant correlation was seen in the left anterior insula (Fig. 2, 24
mm), where there had been little correlation with stimulus tem-
perature. In contrast, the correlations in posterior parietal cortex
and the baseline condition (33C) and averaging across subjects. were greatly reduced. Surprisingly, the extent of the correlated acti-
For example, we found that the 20C stimulus produced signifi- vation in the upper brainstem increased (Fig. 2, 16 mm).
cant rCBF increases on the left (contralateral) side in the mid- Upon closer examination, we noted that the correlations with
dle/posterior insular region, the adjacent lentiform nuclei, posterior stimulus intensity in left middle/posterior and right anterior insular
parietal cortex and middle cingulate cortex and on the right (ipsi- cortex had equal strength. However, the correlation with ratings was
lateral) side in the region of the anterior insula, lentiform nuclei, significantly stronger in right anterior insula than in left middle/pos-
posterior parietal cortex and orbitofrontal cortex (Fig. 2). There terior insula (F-test, F1,116 = 4.61, p < 0.034; Fig. 3). The correlation
was slight activation in the middle/upper brainstem, probably rep- in right anterior insula with ratings was marginally better than with
resenting the periaqueductal gray and the parabrachial nucleus5. temperature (F = 2.1887, p < 0.07, one-tailed). Similarly, activation
The extent of activation in several regions seemed smaller with less in right orbitofrontal cortex was better correlated with ratings than
intense cooling stimuli, so we proceeded to correlational analyses. with temperature (F = 40.3478, p < 108). Given the close corre-
We used a voxel-by-voxel regression analysis to explore cor- spondence of the ratings with stimulus intensity, these are substan-
relations between the intensity of thermal stimulation and tial differences. The significance of these correlations with ratings
regional activity over the entire forebrain, testing the hypotheses was confirmed by ANCOVA with effects due to temperature, inter-
that there would be significant correlations in the left insular subject variability and scan-session factored out (right anterior insu-
region and the other regions noted above. In the left forebrain, la, z = 2.95, p < 0.003; right orbitofrontal, z = 3.59, p < 0.0002). Thus
rCBF was strongly correlated with stimulus intensity in the mid- activation on the right (nondominant) side in the anterior insula
dle/posterior insular region (Fig. 2). There was a weak correla- and orbitofrontal regions (and also left anterior insula) was related
tion in left posterior parietal cortex (but see below). Significant to subjective evaluation of the thermal stimuli.
correlations were observed on the right side in the anterior insu- In contrast, the activation in right and left posterior parietal
lar/lentiform, posterior parietal and orbitofrontal regions and cortices was significantly greater during all thermal stimuli than
middle/upper brainstem. The rCBF correla-
tion with stimulus intensity in the left mid- Table 1. Regression with stimulus temperature.
dle/posterior insula was highly significant and Site Coordinates (x, y, z) Peak Z r Slope p
was the largest in the entire forebrain L. post. insula 36 22 24 4.26 0.55 0.11 0.00005
(p < 0.00005; Fig. 1b; Table 1). We interpret
R. ant. insula 32 12 20 4.12 0.51 0.16 0.00004
this region as the contralateral cortical repre-
sentation of discriminative thermal sensation. brainstem 10 32 16 2.93 0.38 0.10 0.002
We next examined correlations between the R. post. parietal 36 40 36 4.12 0.47 0.12 0.00008
volunteers subjective ratings of thermal stimu- R. orbitofrontal 24 38 0 3.72 0.43 0.12 0.0004
lus intensity and rCBF activity in these regions, Results for the regression against stimulus temperature in the identified regions. Stereotaxic
again using a voxel-by-voxel regression analysis. (Talairach) coordinates are given as x, mediolateral; y, anteroposterior; z, horizontal. The peak Z
As expected, correlations were found in most of value is given for the single voxel at the focus, and the regression values encompass a region extend-
the same regions as with the temperature regres- ing 2 voxels in each direction around the focus (1 voxel for the brainstem site). L., left; R., right.

nature neuroscience volume 3 no 2 february 2000 185


2000 Nature America Inc. http://neurosci.nature.com

articles

in the baseline condition (t-test, p < 105, right Table 2. Regression with raw ratings.
and left), but nearly uniform (ANOVA; right,
F 4,45 = 0.9767, p > 0.42; left, F = 0.4584, Site Coordinates (x, y, z) Peak Z r Slope p
p > 0.77; Fig. 1c). Thus, we interpret these L. post. insula 36 22 24 3.54 0.52 0.13 0.0006
step-like increases in parietal activation as R. ant. insula 32 12 20 4.69 0.60 0.24 0.000004
related to the attention directed to the right Brainstem 10 32 16 2.90 0.41 0.18 0.002
hand during the thermal stimuli. R. post. parietal 36 40 36 2.69 0.47 0.16 0.0005
Frontal projection showed that graded ther- R. orbitofrontal 24 38 0 4.80 0.57 0.21 0.000005
mal activation in the left (contralateral) mid-
dle/posterior insula was focused at the dorsal Results for the regression against subjects ratings in the identified regions. Conventions as in Table 1.
margin (Fig. 4). Notably, because it was centered
in the fundus of the superior limiting (circular) sulcus, the associ- gin of the middle/posterior insula. The activation was linearly
ation of this site with insular cortex might not be appreciated if it correlated with stimulus intensity, indicating that this site repre-
were viewed only in an axial (horizontal) projection. The localiza- sents discriminative thermal sensation. The demonstration that
tion of the foci of activation correlated with the subjects ratings in human thermosensory cortex is located in insular cortex, rather
the right (ipsilateral) anterior insular and orbitofrontal regions is than in parietal somatosensory areas, is significant for several
shown in Fig. 5. reasons. This directly confirms the prediction of several com-
parative and clinical observations, it substantiates the key per-
2000 Nature America Inc. http://neurosci.nature.com

DISCUSSION spective that thermal sensation is a specific aspect of


We found highly significant activation with innocuous cooling enteroception, and it provides strong evidence for the proposal
stimuli at one site in the contralateral forebrain, the dorsal mar- that central pain is an integrative thermosensory disorder.
Innocuous primary afferent (A
cool and C warm) thermoreceptors
innervating skin and deep tissues ter-
20 33 minate in the superficial spinal (and
trigeminal) dorsal horn, where a unique
population of lamina I spinothalamic
neurons is thermoreceptive-specific and
morphologically distinct1619. These neu-
Temp. rons have thresholds near normal skin
regr. temperature, evince linearly graded
responses to innocuous cooling and
have ongoing discharge that is inhibited
by warming and activity that is corre-
lated with discriminative thermal sen-
Rating sation15,16. (Neurons selectively excited
regr.
by warm are also present, though rela-
tively rare.) They project to thalamus in
the crossed lateral spinothalamic tract,
40 mm 32 mm 28 mm 24 mm 20 mm 16 mm
which is clinically and behaviorally crit-
ical for temperature sensibility, as well
as for pain, itch and sexual sensa-
tions2022. In primates, lamina I neurons
20 33
terminate in a dedicated thalamic relay
nucleus (VMpo, the posterior part of
the ventral medial nucleus), which con-
tains thermoreceptive- and nociceptive-
specific neurons2,12. In awake humans,
Temp. such neurons seem to be located in the
regr.
same region, where microstimulation
produces sensations of graded cold or
pain14,23,24. The homologous pathway
exists in cats and rats in primordial
Rating form; VMpo is visible only in primates,
regr. and it is proportionately greatly
enlarged in humans12. Anatomic data in
monkeys indicate that VMpo projects
12 mm 8 mm 0 mm 8 mm 16 mm 20 mm topographically to a cytoarchitectoni-
Fig. 2. Comparison of the statistical maps of rCBF activation for three different analyses at 12 of the 18 axial
cally distinct field in the fundus of the
levels examined. Abbreviations: 20 33, subtraction comparing activation in the 20C stimulus with the superior limiting sulcus at the dorsal
baseline 33C condition; Temp. regr., correlation with stimulus temperature; Rating regr., correlation with margin of middle/posterior insular cor-
subjects ratings. The yellow area indicates activation significant at the z > 2.58 level (p < 0.005), and the red tex5,13. This location matches the site of
area indicates activation significant at the z > 3.90 level (p < 0.0001). Numbers indicate axial (horizontal) level. the human thermosensory cortex

186 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

ceptors1,22. On the premise that temperature is an exteroceptive


Left sensation, prior investigators sought thermoreceptive-specific
insula neurons within the somatosensory system, and the homeostatic
aspect of thermoreception was relegated to the brainstem and
Percent activ-

hypothalamus1. Our findings substantiate a different perspective.


ity

Our findings fit the concept that temperature sensation is a


specific, emergent aspect of enteroception. Like all feelings
from the body, but in contrast to exteroceptive (touch) and
teloreceptive (vision) modalities, thermal sensibility is inher-
ently endowed with a characteristic affect that motivates behav-
ior, and it reflexively generates autonomic responses that signal
its primary role in thermoregulation and its integration with
homeostasis. The functional anatomical characteristics of lam-
ina I and insular cortex manifest this perspective (reviewed in
Right ref. 5). Small-diameter afferents that terminate in lamina I orig-
insula inate from nearly all tissues of the body and are sensitive to
changes in physiological conditions that require systemic
Percent activity

responses (for example, temperature, strong mechanical strain,


2000 Nature America Inc. http://neurosci.nature.com

exercise, low pH, hypoglycemia, hypoxia, hypo-osmolarity, his-


tamine or inflammatory mediators). Consonantly, lamina I has
major projections to spinal autonomic and brainstem homeo-
static integration sites, including regions that receive conver-
gent vagal afferent activity and are heavily interconnected with
the hypothalamus and amygdala, namely, the parabrachial
Ratings nucleus (PB), periaqueductal gray (PAG) and the A1 cell group.
Lamina I also receives descending modulation directly from
Fig. 3. Regression plots comparing the proportional rCBF increases brainstem pre-autonomic sources (A5, A7, raphe) and from the
with ratings in left posterior insula (slope against absolute rCBF = 0.13)
hypothalamus. These and other considerations indicate that the
and right anterior insula (slope against absolute rCBF = 0.24) for each
subject at each temperature.
fundamental role of lamina I is to distribute modality-selective
sensory information on the ongoing physiological status of the
tissues of the body to a hierarchical network involved in the
maintenance of the integrity (or well-being) of the organism5.
demonstrated by the present observations. We conclude that this The termination of the ascending lamina I pathway in insu-
site is the terminus of the lamina I spinothalamocortical path- lar cortex is thus consistent with the view of the insula as limbic
way in humans. Together, these findings demonstrate the central sensory cortex, which is based on its association with visceral
pathway for discriminative thermal sensation.
This observation explains available clinical
findings. Contralateral thermosensory deficits
are caused by lesions involving the lateral
spinothalamic tract, posterolateral thalamus
or parieto-insular cortex6,8,21,25. The critical cor-
tical region is focused at the site identified by
our observations. In contrast, lesions of pari-
etal somatosensory areas have little or no effect
on thermal (or pain) sensation1,25,26. Similarly,
nearly all cooling-sensitive cells recorded in
parietal somatosensory areas show nonlinear
sensitivity associated with the cross-modal
responsiveness of slowly adapting mechanore-

Fig. 4. Localization of thermosensory cortex in


the dorsal margin of left (contralateral) insular cor-
tex (coordinates = 36, 22, 24), identified by
regression analysis of rCBF activation with stimulus
temperature, in frontal, axial and sagittal views. The
low cutoff (purple) of the spectral display is at
z > 2.58 (p < 0.005), and the white indicates z > 3.90
(p < 0.0001). Note that in the axial view, this site
seems parietal simply because the insula cannot be
seen. The activation in right anterior insula is also Temperature regression,
visible in the axial view. These color-coded images left middle posterior insula
were produced with the program Register (David (36, 22, 24)
MacDonald, Montreal Neurological Institute).

nature neuroscience volume 3 no 2 february 2000 187


2000 Nature America Inc. http://neurosci.nature.com

articles

Rating regression, Rating regression,


right anterior insula right orbitofrontal
(32, 12, 20) (24, 38, 0)
2000 Nature America Inc. http://neurosci.nature.com

Fig. 5. Localization of the evaluative regions in right anterior insular and orbitofrontal cortices, identified by regression analysis of rCBF activation
with subjects ratings, in frontal, axial and sagittal views. Conventions as in Fig. 3.

and autonomic function, its multimodal features and, partic- evoked sexual arousal35,3941. Consistent with our association of
ularly, its strong interconnections with hypothalamus, amyg- right anterior insular activity with thermosensory evaluation,
dala and cingulate and orbitofrontal cortices911. Descending similar activation is observed with the signaled anticipation of
projections from insular cortex terminate in lamina I as well as heat pain42 and with tonically evoked heat pain38, in addition to
in the same brainstem pre-autonomic and homeostatic sites contralateral middle/posterior insular activation that can be
noted above27. Stimulation or lesions of insular cortex affect ascribed to the lamina I spinothalamocortical projection from
cardiorespiratory, gastrointestinal, sympathetic and ther- VMpo. Thus, these findings are consistent with the neurological
moregulatory activity11. In primates, the VMpo projection to hypothesis that the right (nondominant) anterior insula is inte-
the dorsal margin of the insula is contiguous anteriorly with gral to mentally generating the image of ones physical state that
the region that receives general (vagal) and special (gustatory) underlies basic emotions43, an essential part of the somatic mark-
visceral input by way of the thalamic VMb nucleus (which is er hypothesis of consciousness, or in different words, a limbic
contiguous with VMpo)27,28. The common source of ascending sensory substrate involved in the evaluation that invests internal
input to insular cortex (by way of VMb) in all mammals is the feelings with emotional significance35. Our findings suggest that
parabrachial nucleus, the brainstem homeostatic site that inte- perceptual dissociation of the affect generated by a thermal stim-
grates both vagal and lamina I inputs; accordingly, the primor- ulus, which depends on thermoregulatory integration, from the
dial role of insular cortex can be regarded as modulation of PB discriminative aspect, which does not44, could occur upon con-
and as a source of multimodal input to goal-directed, homeo- textual evaluation in the right anterior insula, and further in the
static motor processing in the hypothalamus, amygdala and orbitofrontal cortex2,45. Nonetheless, whereas thermal sensation
other sites10,11,29. Consonant with the enormous encephalization (like ticklish or sensual feelings) can be either pleasant or unpleas-
in primates, especially humans, primate enteroceptive sensory ant, thermal distress is selectively associated with concomitant
inputs bypass PB, with a direct gustatory projection from the activation of the anterior cingulate, that is, limbic motor cortex4.
solitary nucleus to VMb30 and a topographic, dedicated lami- The location of human thermosensory cortex corresponds
na I projection to VMpo. These pathways seem to provide a unmistakably with the critical cortical region damaged in certain
highly resolved enteroceptive representation of the bodys con- post-stroke central pain patients8, consistent with the proposal
dition in humans, including the specific sensations of temper- that disruption of thermal sensation may cause central pain5.
ature, pain and other feelings from the body. Head and Holmes recognized that central pain is released by loss
Many observations from functional imaging reinforce these of a specific pain and temperature pathway25, but inferred that
considerations. Middle/posterior insular activation is observed discriminative pain processing normally inhibits the emotional
with gustatory stimulation (taste), fasting (hunger), hyperton- aspect of pain. Others suggest that loss of the lateral pain path-
ic saline injection (thirst and, perhaps, induced hyperthermia), way releases a hypothetical spinoreticulothalamic pathway or that
lactate injection (tachycardia and panic in inducible patients), deafferentation causes bursting and hyperactivity in somatosen-
cholecystokinin injection (tachycardia and induced anxiety), sory pathways7. However, clinical observers report high correla-
inspiration, isometric exercise and various types of cutaneous tion of ongoing, burning central pain with the loss of thermal
and deep pain4,3138. All of these can be considered aspects of pri- sensibility6. Analyses of peripheral nerve block3 and of the ther-
mary enteroceptive sensation. mal grill illusion of pain reveal that reduced thermosensory (cool)
A broader association of anterior insula with internally gen- activity disinhibits a painful burning sensation. In particular, the
erated emotion is suggested by its activation with recall-gener- thermal grill, in which a sensation of burning, ice-like pain is
ated sadness, anticipatory anxiety, panic, disgust and visually generated by innocuous cool (20C) and warm (40C) stimuli

188 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

presented together in a spatially unusual fashion, demonstrates ACKNOWLEDGEMENTS


that reduced activity in the cooling-sensitive lamina I spinothal- We thank S. Goodwin, J. Frost and D. Andrew for technical assistance. This
amocortical pathway unmasks polymodal nociceptive activation study was supported by the Robert S. Flinn Foundation and the Atkinson Pain
of the anterior cingulate cortex that is selectively associated with Research Fund administered by the Barrow Neurological Foundation.
thermal pain4,15. The intense burning experienced when warm
water is applied to a foot numbed by cold is directly comparable RECEIVED 8 OCTOBER; ACCEPTED 6 DECEMBER 1999
to the thermal grill sensation and is an exigent thermoregulato-
ry distress signal. Cutaneous cooling inhibits various enterocep-
tive feelings; one normal function of the cold inhibition of pain 1. Darian-Smith, I. in Handbook of Physiology, Section 1, The Nervous System,
Vol. III, Sensory Processes (ed. Darian-Smith, I.) 879913 (American
may be to produce graded (integrated) differentiation of nox- Physiological Society, Bethesda, 1984).
ious (stressful) cold from innocuous cool, thereby motivating 2. Craig, A. D., Zhang, E. T. & Blomqvist, A. A distinct thermoreceptive
appropriate homeostatic responses to thermal distress that subregion of lamina I in nucleus caudalis of the owl monkey. J. Comp. Neurol.
404, 221234 (1999).
requires protective behavior. 3. Yarnitsky, D. & Ochoa, J. L. Release of cold-induced burning pain by block of
Thus, the thermosensory disinhibition hypothesis proposes cold-specific afferent input. Brain 113, 893902 (1990).
that disruption of the thermosensory (enteroceptive) area in insu- 4. Craig, A. D., Reiman, E. M., Evans, A. & Bushnell, M. C. Functional imaging
of an illusion of pain. Nature 384, 258260 (1996).
lar cortex disinhibits a limbic network, involving brainstem 5. Craig, A. D. A new version of the thalamic disinhibition hypothesis of central
homeostatic sites (PB, PAG) and anterior cingulate cortex, that pain. Pain Forum 7, 114 (1998).
engenders affect associated with thermoregulatory motivation5. 6. Boivie, J., Leijon, G. & Johansson, I. Central post-stroke painA study of the
2000 Nature America Inc. http://neurosci.nature.com

mechanisms through analyses of the sensory abnormalities. Pain 37, 173185


The affective feeling associated with a thermal stimulus reflects (1989).
the needs of thermoregulatory integration44 and seems to persist 7. Pagni, C. A. Central Pain: A Neurosurgical Challenge. (Ediziona Minerva
following thalamocortical thermosensory lesions22. Vague auto- Medica S. P. A., Turin, 1998).
8. Schmahmann, J. D. & Leifer, D. Parietal pseudothalamic pain syndrome:
nomic disturbances are repeatedly noted in central pain patients, Clinical features and anatomic correlates. Arch. Neurol. 49, 10321037
and many experience relief simply by going into a warm room, (1992).
9. Mesulam, M.-M. & Mufson, E. J. Insula of the old world monkey. III: Efferent
exercising lightly or sitting in a spa6,25. The present results pro- cortical output and comments on function. J. Comp. Neurol. 212, 3852
vide strong evidence supporting the idea that central pain is an (1982).
enteroceptive, thermoregulatory dysfunction. 10. Allen, G. V., Saper, C. B., Hurley, K. M. & Cechetto, D. F. Organization of
visceral and limbic connections in the insular cortex of the rat. J. Comp.
Neurol. 311, 116 (1991).
11. Augustine, J. R. Circuitry and functional aspects of the insular lobe in
METHODS primates including humans. Brain Res. Rev. 22, 229244 (1996).
Our protocol was approved by local institutional review boards. We mea- 12. Craig, A. D., Bushnell, M. C., Zhang, E.-T. & Blomqvist, A. A thalamic
sured rCBF during 12 scans (60 s each) in each of 10 normal right-hand- nucleus specific for pain and temperature sensation. Nature 372, 770773
ed volunteers (3 female, 7 male; ages 2349, mean 33) using an ECAT 951/31 (1994).
13. Craig, A. D. in Forebrain Areas Involved in Pain Processing. (eds. Besson, J.-M.,
scanner (CTI, Knoxville, Tennessee). After a 30-min transmission scan, Guilbaud, G. & Ollat, H.) 1326 (John Libbey, Paris, 1995).
bolus injections of 40 mCi of H215O were made at 15-min intervals4. Ther- 14. Davis, K. D. et al. Thalamic relay site for cold perception in humans.
mal stimuli were applied to the palmar surface of the right hand with a 20 J. Neurophysiol. 81, 19701973 (1999).
14 cm feedback-controlled Peltier device4,15. The volunteers had received 15. Craig, A. D. & Bushnell, M. C. The thermal grill illusion: unmasking the burn
of cold pain. Science 265, 252255 (1994).
similar, but not identical, stimuli before the scans. The cooling stimuli
16. Dostrovsky, J. O. & Craig, A. D. Cooling-specific spinothalamic neurons in
ramped at 1 per s from a baseline of 33C and attained a stable plateau the monkey. J. Neurophysiol. 76, 36563665 (1996).
temperature of 30, 28, 26, 24 or 20C before each bolus injection, whereas 17. Christensen, B. N. & Perl, E. R. Spinal neurons specifically excited by noxious
the neutral condition remained at 33C. Each volunteer was instructed to or thermal stimuli: marginal zone of the dorsal horn. J. Neurophysiol. 33,
remain still with eyes closed. Scanning was initiated upon the sharp increase 293307 (1970).
18. Dostrovsky, J. O. & Hellon, R. F. The representation of facial temperature in
in measured radioactivity (1518 seconds after the injection). The temper- the caudal trigeminal nucleus of the cat. J. Physiol. (Lond.) 277, 2947
ature was stable through the first 30 s of the scan and then returned to base- (1978).
line (ramp rate, 10 per s), to maximize the contrast between 19. Han, Z.-S., Zhang, E.-T. & Craig, A. D. Nociceptive and thermoreceptive
state-dependent rCBF increases and noise46. The volunteers gave a magni- lamina I neurons are anatomically distinct. Nat. Neurosci. 1, 218225 (1998).
tude estimate of the perceived intensity of cold following each scan using 20. White, J. C. & Sweet, W. H. Pain and the Neurosurgeon: A Forty-Year
Experience 1000 (Thomas, Springfield, Massachusetts, 1969).
an open-ended scale of zero to ten. The six different stimulus conditions 21. Norrsell, U. Behavioural thermosensitivity after unilateral, partial lesions of
were presented two times each, first in a descending sequence of tempera- the lateral funiculus in the cervical spinal cord of the cat. Exp. Brain Res. 78,
tures and then in an ascending sequence, to counterbalance order effects. 369373 (1989).
The scanner produced 31 horizontal 128 128 slices with a separa- 22. Norrsell, U. & Craig, A. D. Behavioral thermosensitivity after lesions of
thalamic target areas of a thermosensory spinothalamic pathway in the cat.
tion of 3.375 mm, a 10.8-cm axial field of view, an in-plane resolution J. Neurophysiol. 82, 611625 (1999).
of 9.5 mm, full width at half maximum (FWHM) and an axial resolu- 23. Dostrovsky, J. O., Wells, F. E. B. & Tasker, R. R. in Processing and Inhibition of
tion of 5.07.1 mm FWHM. The data were analyzed with Statistical Para- Nociceptive Information, International Congress Series 989 (eds. Inoka, R.,
metric Mapping (SPM96) implemented in Matlab (Mathworks, Sherborn Shigenaga, Y. & Tohyama, M.) 115120 (Excerpta Medica, Amsterdam,
Massachusetts)47,48. The PET images for each subject were aligned with 1992).
24. Lenz, F. A. et al. Neurons in the area of human thalamic nucleus ventralis
the initial scan49, transformed into a standard stereotaxic coordinate caudalis respond to painful heat stimuli. Brain Res. 623, 235240 (1993).
space50, smoothed with an isotropic Gaussian kernel (16 mm FWHM) 25. Head, H. & Holmes, G. Sensory disturbances from cerebral lesions. Brain 34,
and normalized for global variations in absolute measurements. Statistical 102254 (1911).
parametric maps were constructed using data from all ten volunteers and 26. Adams, R. W. & Burke, D. Deficits of thermal sensation in patients with
unilateral cerebral lesions. Electroencephalogr. Clin. Neurophysiol. 73,
superimposed onto a spatially standardized MRI volume. The statistical 443452 (1989).
comparisons between different stimulus conditions and the regression 27. Yasui, Y., Breder, C. D., Saper, C. B. & Cechetto, D. F. Autonomic responses
analyses were performed at each voxel using a multi-subject design with and efferent pathways from the insular cortex in the rat. J. Comp. Neurol. 303,
replications. Normalized t-values (z-scores) were computed to compare 355374 (1991).
differences between each active thermal condition and the neutral con- 28. Pritchard, T. C., Hamilton, R. B., Morse, J. R. & Norgren, R. Projections of
thalamic gustatory and lingual areas in the monkey, Macaca fascicularis.
dition and for regressions against temperature and magnitude ratings. J. Comp. Neurol. 244, 213228 (1986).
Significant rCBF activation in the regions of interest occurred at z > 2.58, 29. Shi, C. J. & Cassell, M. D. Cortical, thalamic, and amygdaloid connections of the
yielding p < 0.005 uncorrected for multiple comparisons35,38. anterior and posterior insular cortices. J. Comp. Neurol. 399, 440468 (1998).

nature neuroscience volume 3 no 2 february 2000 189


2000 Nature America Inc. http://neurosci.nature.com

articles

30. Beckstead, R .M., Morse, J. R. & Norgren, R. The nucleus of the solitary tract 41. Mayberg, H. S. et al. Reciprocal limbic-cortical function and negative mood:
in the monkey: projections to the thalamus and brain stem nuclei. J. Comp. converging PET findings in depression and normal sadness. Am. J. Psychiatry
Neurol. 190, 259282 (1980). 156, 675682 (1999).
31. Kinomura, S. et al. Functional anatomy of taste perception in the human brain 42. Ploghaus, A. et al. Dissociating pain from its anticipation in the human brain.
studied with positron emission tomography. Brain Res. 659, 263266 (1994). Science 284, 19791981 (1999).
32. Benkelfat, C. et al. Functional neuroanatomy of CCK4-induced anxiety in normal 43. Damasio, A. R. Descartes Error: Emotion, Reason, and the Human Brain 312
healthy volunteers. Am. J. Psychiatry 152, 11801184 (1995). (Putnam, New York, 1993).
33. Casey, K. L., Minoshima, S., Morrow, T. J. & Koeppe, R. A. Comparison of 44. Mower, G. Perceived intensity of peripheral thermal stimuli is independent
human cerebral activation patterns during cutaneous warmth, heat pain, and of internal body temperature. J. Comp. Physiol. Psychol. 90, 11521155
deep cold pain. J. Neurophysiol. 76, 571581 (1996). (1976).
34. King, A. B., Menon, R. S., Hachinski, V. & Cechetto, D. F. Human forebrain 45. Francis, S. et al. The representation of pleasant touch in the brain and its
activation by visceral stimuli. J. Comp. Neurol. 413, 572582 (1999). relationship with taste and olfactory areas. Neuroreport 10, 453459 (1999).
35. Reiman, E. M. The application of positron emission tomography to the study 46. Volkow, N. D., Mullani, N., Gould, L. K., Adler, S. S. & Gatley, S. J. Sensitivity
of normal and pathological emotions. J. Clin. Psychiatry 58, 412 (1997). of measurements of regional brain activation with oxygen-15-water and PET
36. Denton, D. et al. Neuroimaging of genesis and satiation of thirst and an to time of stimulation and period of image reconstruction. J. Nucl. Med. 32,
interoceptor- driven theory of origins of primary consciousness. Proc. Natl. 5861 (1991).
Acad. Sci. USA 96, 53045309 (1999). 47. Friston, K. J., Frith, C. D., Liddle, P. F. & Frackowiak, R. S. Comparing
37. Tataranni, P. A. et al. Neuroanatomical correlates of hunger and satiation in functional (PET) images: the assessment of significant change. J. Cereb. Blood
humans using positron emission tomography. Proc. Natl. Acad. Sci. USA 96, Flow Metab. 11, 690699 (1991).
45694574 (1999). 48. Worsley, K. J., Evans, A. C., Marrett, S. & Neelin, P. A three-dimensional
38. Derbyshire, S. W. G. & Jones, A. K .P. Cerebral responses to a continual tonic statistical analysis for CBF activation studies in human brain. J. Cereb. Blood
pain stimulus measured using positron emission tomography. Pain 76, Flow Metab. 12, 900 918 (1992).
127135 (1998). 49. Woods, R. P., Cherry, S. R. & Mazziotta, J. C. Rapid automated algorithm for
39. Stoleru, S. et al. Neuroanatomical correlates of visually evoked sexual arousal aligning and reslicing PET images. J. Comput. Assist. Tomogr. 16, 620633
2000 Nature America Inc. http://neurosci.nature.com

in human males. Arch. Sex. Behav. 28, 121 (1999). (1992).


40. Phillips, M. L. et al. A specific neural substrate for perceiving facial 50. Talairach, J. & Tournoux, P. Co-Planar Stereotaxic Atlas of the Human Brain
expressions of disgust. Nature 389, 495498 (1997). (Thieme, New York, 1988).

190 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Expertise for cars and birds recruits


brain areas involved in face
recognition
Isabel Gauthier1,2, Pawel Skudlarski2, John C. Gore2 and Adam W. Anderson2

1 Present Address: Department of Psychology, Vanderbilt University, Wilson Hall, Nashville, Tennessee 37240, USA
2 Department of Diagnostic Radiology, Yale University Medical School, Fitkin Basement, 333 Cedar Street, New Haven, Connecticut 06510, USA
Correspondence should be addressed to I.G. (isabel.gauthier@vanderbilt.edu)

Expertise with unfamiliar objects (greebles) recruits face-selective areas in the fusiform gyrus (FFA)
2000 Nature America Inc. http://neurosci.nature.com

and occipital lobe (OFA). Here we extend this finding to other homogeneous categories. Bird and
car experts were tested with functional magnetic resonance imaging during tasks with faces,
familiar objects, cars and birds. Homogeneous categories activated the FFA more than familiar
objects. Moreover, the right FFA and OFA showed significant expertise effects. An independent
behavioral test of expertise predicted relative activation in the right FFA for birds versus cars within
each group. The results suggest that level of categorization and expertise, rather than superficial
properties of objects, determine the specialization of the FFA.

Face and object recognition differ in at least two ways. First, faces activation in the FFA when matched to specific labels as compared
are recognized at a more specific level of categorization (for exam- to more categorical ones (for example, ketchup bottle versus bot-
ple, Adam) than most objects (for example, chair or car). Sec- tle)3,30. Third, expertise with animal-like unfamiliar objects (gree-
ond, although we are experts with faces, we have much less bles) recruits the right FFA4. However, it remains unclear whether
experience discriminating among members of other categories. expertise with any homogeneous category is capable of recruiting
Level of categorization and expertise are relevant even for unfa- the neural substrate of face recognition.
miliar faces and objects. A person passed on the street may be This experiment had three purposes. First, we tested whether
encoded at the individual level and recognized the next day, long-term expertise with birds and cars would recruit face-selec-
whereas a mug may be replaced by another mug without our tive areas. Second, the interaction between level of categoriza-
noticing. Processing biases for different categories depend on our tion and expertise was investigated. Third, we tested how these
experience with levels of categorization and our expertise in two factors depend on attention to stimulus identity. The FFA
extracting diagnostic features1. typically activates more for faces than objects, even during passive
Viewing faces activates a small extrastriate region called the viewing7. This suggests that faces are processed automatically at
fusiform face area (FFA)210. Neuropsychological studies suggest the subordinate level. Here we asked whether this is also true for
that the brain areas responsible for face and object processing other expertise domains.
can be dissociated1114. According to one view, extrastriate cor-
tex contains a map of visual features1516, suggesting that the same RESULTS
region should not be recruited for processing different object cat- We tested 11 car experts and 8 bird experts with many years of
egories when the relevant features differ. experience recognizing car models or bird species (Table 1). The
On the other hand, prosopagnosia is often associated with right and left FFA and right occipital face area (OFA) were
deficits discriminating among nonface objects within categories. defined in passive-viewing localizer scans (see Methods). The
For example, a bird watcher became unable to identify birds17, OFA is also face selective31 and active in greeble experts4. A right
whereas another patient could no longer identify car makes18. FFA was found in all subjects (median size, 6 voxels), a left FFA
Thus, one hypothesis holds that prosopagnosia is a deficit in was found in 13 subjects (4 bird experts, 9 car experts; median
evoking a specific context from a stimulus belonging to a class size, 5 voxels) and a right OFA in 15 subjects (7 bird experts, 8
of visually similar objects19. At least some prosopagnosic patients car experts; median size, 7 voxels).
have difficulty with classes in which objects are both visually and Subjects also underwent identity and location scans. Stimulus
semantically homogeneous20,21. Evidence from brain-lesion stud- presentation was identical in both conditions, and subjects detect-
ies is still under debate13,22; however, additional data from brain ed immediate (1-back) repetitions in either the identity of the pic-
imaging may help resolve these questions. ture or its location while ignoring the other dimension. Blocks of
Several lines of research converge to suggest that level of cate- 16 grayscale faces, objects, cars or birds shown sequentially were
gorization and expertise account for a large part of the activation alternated with periods of fixation (Fig. 1). Pilot experiments indi-
difference between faces and objects. First, behavioral effects2325 cated that the absence of color cues did not eliminate the advantage
once thought unique to faces have been obtained with objects, of experts over novices. Behavioral data in the scanner was available
often with expert subjects2629. Second, nonface objects elicit more for 16 of the 19 subjects. Performance was better in the identity

nature neuroscience volume 3 no 2 february 2000 191


2000 Nature America Inc. http://neurosci.nature.com

articles

Inspection of Fig. 2 suggests baseline differences between cate-


gories and between groups. First, responses to birds were larger than
to cars in FFAs of both hemispheres (right, F1,17 = 11.13, p < 0.05;
left, F1,11 = 5.47, p < 0.05). Again, although animal faces may acti-
vate this area more than objects10, here we found no difference
between cars and birds in novices. Bird experts showed more acti-
vation for any object category than car experts, although not signif-
icantly in any ROI. Given these baseline trends, it was crucial to
measure the effect of expertise by comparing activation for cars and
birds in the two groups. The predicted expertise effect was signifi-
cant (a group category interaction) in the right FFA (Figs. 2 and 3;
F1,17 = 19.22, p < 0.0005) and in the right OFA (F1,13 = 4.86, p < 0.05).
There was no expertise effect in the left FFA (F < 1).
One important question is whether this expertise effect aris-
es from the same area as face expertise. To test this, we used a set
a of criteria (see Methods) based on a definition of face cells in neu-
rophysiology32. This defines a smaller FFA than any other defini-
tion to our knowledge (also eliminating the majority of OFA and
2000 Nature America Inc. http://neurosci.nature.com

left FFA ROIs, which were not analyzed further). We call this ROI
the center of the FFA (median = 3 voxels), in which each voxel is
highly face selective. Even in this ROI, both the level of catego-
rization effect in novices (F1,17 = 6.37, p < 0.02) and the expertise
effect were present (F1,17 = 10.25, p < 0.006; Fig. 3). These effects
also held when analyzed in a subset of subjects whose FFA could
Location Location Identity be defined using described criteria6,10 (see supplemental material
or identity
at http://neurosci.nature.com/web_specials/). To assess the mag-
Time
b nitude of the expertise effect, we plotted the main effects and inter-
action separately for the center of the right FFA33 (Fig. 4). The
Fig. 1. Examples of stimuli and tasks for the fMRI protocol. (a) Images statistically significant expertise effect contributed a difference of
(256 256 pixels in size, 256 grays) from each of 4 categories about 0.4% signal change between groups, whereas the group and
(Caucasian faces without hair, passerine birds from New England, car category main effects contributed about 0.3% and 0.1% signal
models for the years 1995 and 1998 and various familiar objects) were change, respectively, and were not significant (F < 1 for both).
used in the fMRI study. (b) Example of stimulus presentation during the Corresponding values in the larger right FFA ROI were 0.2, 0.1
fMRI runs. Subjects made 1-back repetition judgments regarding either
and 0.3, respectively. The expertise effect alone accounted for 32%
location or identity (an identity repeat would show identical images,
although sometimes in different locationssee Methods for details). of the difference between faces and objects in the right FFA defined
at t = 2 and for 36% in the center of the right FFA.
We measured the center of mass of the signal change for acti-
vated voxels for birds, cars and faces (relative to objects). This
than the location runs (identity performance s.e., 89.4 2.1; was done in a ROI of 25 25 voxels (each 1.3 mm by 1.7 mm,
location, 86.0 2.3; F1,14 = 9.98, p < 0.01) and this effect was larg- y x over 3 slices in Talairach space, centered on the right and
er for birds and objects than for cars and faces (task category left FFA from the localizer). The only significant differences for
interaction, F1,14 = 5.86, p < 0.01). These categories varied more in cars or birds relative to faces were obtained in novice subjects
shape, making location judgments more difficult.
The percent signal changes in the three regions of
interest (ROIs) were assessed using a fixation baseline. Table 1. Subject information and behavioral results.
First, we describe all significant effects pooled across Bird experts Car experts
task, coming back to this factor later. The level of cat- Mean age s.e. 34.4 2.0 31 2.5
egorization effect was measured by comparing activa- Mean years experience se 18 3.3 20.6 3.8
tion in novices to cars or birds versus objects. The
effect of level of categorization was significant in the
right FFA (F1,17 = 14.36, p < 0.02) and in the left FFA Behavioral data during fMRI
(F1,11 = 8.76, p = 0.02.). This effect was marginal in the (% correct identity s.e.; location s.e.)
right OFA (F1,13 = 3.67, p < 0.08). The interaction Objects 86 3; 81 4 93 3; 88 3
between level and group was significant in both the Faces 85 3; 82 3 92 3; 91 3
right FFA (F 1,17 = 6.61, p < 0.02) and the left FFA Cars 84 3; 81 3 92 2; 91 2
(F1,11 = 6.47, p < 0.05). Post-hoc tests (p < 0.05) indi- Birds 87 3; 81 4 92 3; 89 3
cated that the level effect was only significant for car
experts viewing birds. It may be tempting to believe Behavioral data pre-test (d s.e.)
that birds activate the FFA because of their faces10. Birds upright 2.53 0.10 1.06 0.07
However, the difference between birds and cars for
Cars upright 1.41 0.12 2.42 0.14
novices was not significant in either area (p > 0.5 for
Birds inverted 2.23 0.20 1.01 0.09
both), and the group effect arises from a difference in
activity for common objects (larger in birders). Cars inverted 0.84 0.13 1.58 0.20

192 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

bird experts
3.125 7 mm3) window in Fig. 5.
Experts (and to some extent bird-
car experts ers viewing cars) showed a distri-
bution of activation for birds and
Right FFA Center of right FFA
cars that was relatively limited to
the localizer peak of the FFA. The
mean percent signal change in the
Mean % signal change

Mean % signal change


center voxel was compared to that
in the 8 voxels surrounding it, and
to the surrounding outside 16
voxels. An ANOVA (3 regions 3
categories 2 groups) revealed a
main effect of region (F2,22 = 7.18,
p < 0.004) with more activity in
the center than in both outer
regions. Because of greater activ-
Objects Cars Birds Faces Objects Cars Birds Faces
ity for faces than birds and cars
only in the center, the category
2000 Nature America Inc. http://neurosci.nature.com

region interaction was marginal


Left FFA Right OFA
(F4,44 = 2.18, p < 0.09), consistent
with our other analyses. Crucial-
ly, there was no significant differ-
ence among the three categories
Mean % signal change

Mean % signal change

in activity for each of the two sur-


rounding regions, suggesting that
activation is as focused for objects
as for faces.
As a more direct way of assess-
ing the expertise effect, we mea-
sured the correlation between
behavioral performance outside
Objects Cars Birds Faces Objects Cars Birds Faces the scanner with the signal
change in the three ROIs during
Fig. 2. Mean percent signal change for each object category in the two expert groups in three face-specific ROIs the location and identity tasks. In
and in the center of the right FFA. The average percent signal increase from fixation for each object category in the
the behavioral test, subjects
different ROIs was averaged across subjects in each ROI for each expert group. Error bars indicate standard error
of the mean. The Talairach coordinates for the center of each ROI standard error were right FFA, x = 38 2, judged whether sequentially pre-
y = 50 1, z = 7 1; left FFA, x = 38 2, y = 56 4, z = 6 2; right OFA, x = 40 2, y = 75 3, z = 3 1. sented pairs of birds and cars
(upright or inverted) belonged to
the same species or car model.
The expertise effect was signifi-
(see Table 2). Center of mass for expert categories was indistin- cant (group category interaction; Table 1), bird experts being
guishable from that obtained for faces (even using a lenient sta- more sensitive for birds than cars and vice versa for car experts
tistical test). We also compared the activation distribution in the (F1,17 = 59.40, p < 0.0001). The effect of orientation and interac-
right FFA for the three categories by averaging nonspatially tion of category with orientation were significant, with the inver-
smoothed individual maps, centered on the most face-selective sion effect stronger for cars than for birds (F1,17 = 14.27, p <
voxel in the localizer. This is shown in a 5 5 voxel (each 3.125 0.002). Both groups were poorer with inverted than upright cars,

Fig. 3. The right FFA shows an expertise


Faces objects Cars objects Birds objects
effect for birds and cars. One axial oblique
slice through the FFA for one expert for each
category shows the t-maps obtained when
Car expert

comparing the activation for faces, cars and


birds with the activation elicited by objects t = 4.5
during the location 1-back runs. The voxels
marked by white crosses indicate the right
FFA and OFA as defined in the passive viewing
runs for these two subjects. (In this car
expert, the OFA was actually in the slice
immediately below and is shown on the same
Bird expert

slice as the FFA only to illustrate its in-plane


location.) Note that the center of the right
t=1
FFA may be slightly different depending on the
task (here passive viewing versus 1-back loca-
tion) and that its size varies between subjects.

nature neuroscience volume 3 no 2 february 2000 193


2000 Nature America Inc. http://neurosci.nature.com

articles

whereas the inversion effect for birds only bird experts


approached statistical significance in bird- Main effects car experts Interaction
ers (p = 0.068).
A group analysis was performed on
the fMRI data during all 1-back tasks

Percent signal change

Percent signal change


with cars and birds (see Methods). This
less precise analysis allowed us to seek
other regions showing an expertise effect
regardless of the category, beyond the
ones we could define functionally. This
showed an area in right ventral temporal
cortex that was more activated in experts
than novices (Fig. 6). In addition to this
stream of activation, going from the right
OFA toward the right FFA, a bilateral
region in the parahippocampal gyrus was Cars Birds Cars Birds
also more active in experts. This area
overlaps with the parahippocampal place Fig. 4. Main effects with grand mean and interaction partialed out and interaction effect with the
2000 Nature America Inc. http://neurosci.nature.com

area (PPA)34, functionally defined as the grand mean and main effects partialed out, in the center of the right FFA. Each observed condition
region responding more to scenes than mean can be reconstructed by adding the value for the main effects and the interaction effect to the
objects. (It also responds more to objects grand mean (in this case, 0.845).
than faces.) Further work will be required
to identify the role of this area in percep-
tual expertise. The only region more acti-
vated in novices than experts was a small bilateral area of the small (1-back identity versus passive viewing; but see ref. 36). We
lateral occipital gyrus, superior to the OFA. This area has been also found no effect of task on the advantage of faces over objects in
found to activate more for letter strings than faces35, and its selec- the right FFA (p > 0.28), nor on the expertise effect. Expertise may
tive activation for novices could reflect a switch from a featural influence how objects are automatically processed, an idea that we
to a more configural strategy. come back to in our discussion.
In each ROI, we correlated the percent signal change for birds
minus cars with relative expertise, the difference in sensitivity DISCUSSION
(d) for upright birds minus upright cars. As the 2 groups com- Previous studies suggest that level of categorization and expertise
bined would produce a bimodal distribution, the correlation coef- contribute to the specialization of the FFA. The present results show
ficients were calculated for each group separately using our largest how their contributions add up to account for a considerable part
homogeneous sample (the 12 subjects scanned with axial slices.) of the difference typically found between objects and faces.
For both groups, relative expertise was positively correlated with In our experiment, experts would know more names for the
relative percent signal change for birds versus cars in the right birds or cars than novices would. However, naming is not likely
FFA and only for the location task (car experts, r = 0.75; bird to account for the effects in the FFA because unfamiliar faces
experts, r = 0.82; p < 0.05 for both; Fig. 7). activated this area the most, whereas common objects that are
We also considered task effects beyond that found in the corre- easily named elicited the least activation. In addition, expertise
lation analyses. The only ROI showing a significant influence of effects for novel objects can be obtained in the FFA for unfa-
task was the right FFA, where this factor interacted with level of cat- miliar exemplars of a trained category4.
egorization (F1,17 = 6.58, p < 0.02): the subordinate-level advantage Why would faces recruit the FFA more than expert recog-
was larger when novices attended to the identity than to the location nition of objects? There are many possibilities. First, the FFA
of the stimuli. In prior studies6,10, the effect of task in the FFA was may be dedicated to face recognition (innately or through expe-
rience), although it may mediate the processing of
Table 2. Center of mass coordinates in the middle temporal lobe for other objects to some extent. At the least, our study
category-selective areas, given in Talairach coordinates. demonstrates that an innate bias is unnecessary for
Left hemisphere Right hemisphere objects to recruit this area with expertise. Second,
x y z x y z we cannot claim to have equated objects with faces
on level of categorization and expertise22. The faces
Bird experts
may constitute a more visually homogeneous set
Faces 31.3 49.8 7.6 40.8 48.2 8.5
than our bird or car images. Faces are recognized
Cars 29.3* 47.3* 7.8 39.9 47.5 9.0 as individual exemplars, whereas even experts
Birds 30.8 49.6 7.9 40.3 48.1 8.3 mainly recognize cars and birds at the
model/species level. Although our subjects had
Car experts years of experience with cars or birds, they still had
Faces 29.0 48.7 9.1 38.6 48.1 9.1 been practicing face recognition for many more
Cars 28.9 49.2 8.1 38.3 47.2 8.5 years. Thus, face recognition being in a sense more
Birds 30.5* 49.8 7.0 41.2* 47.8 8.9 subordinate and relying on greater expertise may
be what make it seem special, leaving little contri-
*Value significantly different from the coordinate for faces in the same expert group accord- bution for a component of object category per se.
ing to a least significant difference test; p < 0.05.
Additionally, categorization level and expertise may

194 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

Fig. 5. Spatial distribution of percent signal Mean % signal change


change for faces, birds and cars (relative to an
0 0.9 1.8
objects baseline) in a 5 5 voxel window in
the right FFA, centered on the most strongly
activated voxel in the localizer. Dashed lines
indicate the three regions within which acti- Faces objects Birds objects Cars objects
vation was averaged for analyses. Note that
the highest activation during experimental
runs for faces may not be identical to the
highest peak in the localizer (consider car

Bird experts
experts in the faces objects condition).

be only two of several factors that


determine the specialization of this
area. (Other factors may include sym-
metry, properties of associated seman-
tic knowledge, number of exemplars,
2000 Nature America Inc. http://neurosci.nature.com

value to the perceiver37.)


Car experts

The effect obtained in the right


OFA suggests that expertise may be
responsible for specialization of a large
part of the face recognition system (at
least in the right hemisphere). In the
left FFA, we found an effect of level of
categorization, with no detectable con-
tribution of expertise. Whereas subordinate-level processing There seems to be an important interaction between auto-
may recruit both hemispheres, here visual expert recognition maticity of processing at the subordinate level and expertise. It is
of homogeneous categories seems to be mainly a right hemi- argued that the preference of the FFA for faces does not depend on
sphere process. the task6,10 (but see ref. 35), a claim also supported by our results.
Our most striking result may be a very strong correlation However, we found an interaction between level of categorization
between a behavioral test of object expertise and the relative acti- and task for novices, indicating that, for most people, simply seeing
vation in the right FFA for birds and cars. It is remarkable that an object among similar exemplars may not prompt complete sub-
the expertise of a subject was so accurately predicted from the ordinate-level processing. Automatic subordinate-level processing
activation in a small part (six voxels) of the brain, especially as for experts could also explain a surprising finding: the correlation
the behavioral and fMRI experiments shared neither a common between behavior and activation in the right FFA was significant
task nor stimulus set. In addition, this analysis suggests that acti- only when subjects attended to the location of the objects. During
vation of the right FFA was more directly correlated with expert the identity task, subjects had to perform subordinate-level recog-
performance than the right OFA. nition with both categories, regardless of expertise. Novices may

Fig. 6. Expertise effect in the temporal cortex. The t-maps


for all subjects with axial slices (14) were transformed into a
common standard space. Voxels showing a significant exper-
tise effect across subjects (p < 0.01) are displayed on the
transformed anatomical images for slices 2 to 5 for a single
subject. The red to yellow voxels were more active for
experts than novices across the identity and location tasks,
whereas the blue voxels were more active for novices. The
right hemisphere is shown on the left. The FFA is typically
found in slice three.

nature neuroscience volume 3 no 2 february 2000 195


2000 Nature America Inc. http://neurosci.nature.com

articles

Location task tral pathway, because it suggests that responses of neurons in


extrastriate cortex may not be organized according to the visu-
bird advantage

al features that they detect15,16; rather, their functional organi-


car experts, r = 0.75*
bird experts, r = 0.82* zation may depend on the different processes important for
Relative % signal change

object recognition. For instance, some areas may be more suit-


ed for featural processing, whereas other areas may support
configural and holistic processing, hallmarks of subordinate-
level expertise. Our results suggest that expert subordinate-level
recognition for any category may be mediated in the same
regions, either by virtue of activating common cells or through
selectively activating different populations that are intermin-
gled. Other techniques, such as single-cell recording, will be
car advantage

necessary to distinguish between these two alternatives.


METHODS
Subjects. Subjects, all male, included 11 car experts and 8 bird experts.
Informed consent was obtained from each subject, and the study was
Relative expertise (d) approved by the Human Investigation Committee at the School of Med-
2000 Nature America Inc. http://neurosci.nature.com

car advantage bird advantage icine, Yale University. Eight subjects were left handed. Handedness did
not correlate with any effect reported here.

Stimuli. One hundred and seventy six images each of passerine birds and
cars were obtained from public sources on the world-wide web. Images
Identity task were converted to 8-bit grayscale 256 256-pixel format, and objects
were isolated and placed on a 50% gray background. Objects were select-
ed to be familiar to our expert population. For each category, 112 images
bird advantage

were used in the behavioral test, whereas the remaining 64 objects from
car experts, r = 0.10
bird experts, r = 0.004
each category were used for experimental scans. Faces without hair
(n = 64, scanned in a 3-D laser scanner, courtesy of Niko Troje and Hein-
Relative % signal change

rich Blthoff, Max Planck Institute, Tbingen, Germany) and 64 images


of non-living familiar objects were prepared in the same way as the cars
and birds and also used in experimental scans. Localizer scans used 90
grayscale photographs of faces and 90 pictures of familiar objects.

Behavioral task. Each subject performed 10 blocks of 56 sequential


matching trials, alternating between blocks showing birds or cars. There
were four conditions (upright and inverted cars and birds). Each trial
car advantage

showed two images from the same category and orientation. Upright and
inverted trials were randomly intermixed. On each trial, a fixation cross
appeared for 500 ms, followed by stimulus 1 for 1000 ms and a pattern
mask for 500 ms before stimulus 2 appeared and remained on the screen
until a response was made. Subjects judged whether the two images
showed birds from the same species or whether cars were from the same
Relative expertise (d)
model but different years (mostly 1995 versus 1998). No difference was
car advantage bird advantage found in mean sensitivity between categories for novices. However,
responses to cars were slower than responses to birds for all subjects (RTs
Fig. 7. Relationship between a behavioral measure of expertise and for hits with cars, 1138 ms; birds, 1046 ms; p < 0.05, suggesting that the
activation in the right FFA. Relative expertise is the sensitivity (d) for cars were more difficult).
bird minus car matching. The dashed and full lines respectively indicate
the best linear fits for car and bird experts. Significant correlation coef- fMRI task. Experimental scans consisted of three runs of a one-back loca-
ficients are marked with an asterisk (p < 0.05). tion task alternated with three runs of a one-back identity task. The only
difference between identity and location runs was instructions to detect
immediate repetitions in either location or identity (Fig. 1). Each run
lasted 5 min, 36 s and consisted of 16 epochs (16 s each) with 5 fixation
then use a featural strategy, whereas experts may use a more con- periods (16 s each) interleaved at regular intervals. During each epoch, 16
figural strategy2628. Perhaps only configural processing is a good objects appeared, each shown for 725 ms followed by a 275 ms blank.
predictor of behavioral expertise. In contrast, during the location Objects (each 12 12) appeared in one of 8 locations within an overall
task novices may not access the subordinate level, whereas experts area subtending 18 18 of visual angle. The order of the four categories
did so automatically. was counterbalanced across runs.
Birds and cars differ in many aspects. (Birds are small ani-
mals with moveable parts, covered with feathers that have spe- ROI selection. Regions of interests were functionally defined using two
localizer scans, which included 16 epochs (16 s each) of passive viewing of
cific markings; cars are large man-made objects made of metal
faces or common objects centered on the screen (25 pictures per epoch).
and typically uniform in texture.) Combined with a previous Each run began with 16 s of fixation, and an 8-s fixation period was includ-
study showing an expertise effect in the FFA with greebles4, ed after every 2 passive viewing epochs. The right and left FFA and right
our results suggest very few constraints on the structure of the OFA were defined as contiguous voxels activated at an arbitrary threshold of
objects for which expertise can recruit this small area. This is t = 2 in the middle fusiform gyri (cd, F-G, 9-10 in Talairach space), and
important for any theory of visual representation in the ven- the same threshold was applied in the right ventral occipital lobe (c-d, H-I,

196 nature neuroscience volume 3 no 2 february 2000


2000 Nature America Inc. http://neurosci.nature.com

articles

9-10) for the right OFA . The raw data were noisier than when using a high- 3. Gauthier, I., Tarr, M. J., Moylan, J., Anderson, A. W. & Gore, J. C. The functionally-
er field scanner and a surface coil6,10 so the same level of significance for defined face area is engaged by subordinate-level recognition. Cognit.
ROI definitions could not be applied. However, the magnitude and spatial Neuropsychol. (in press).
4. Gauthier, I., Tarr, M. J., Anderson, A. W., Skudlarski, P. & Gore, J. C. Activation of
extent of the effect for a given functional area should be similar regardless of the middle fusiform face area increases with expertise in recognizing novel objects,
statistical power, and we used a criterion leading to ROIs comparable in Nat. Neurosci. 2, 568573 (1999).
size to those in published studies6,10 . Our FFAs show at least twice as much 5. Haxby, J. V. et al. The functional organization of human extrastriate cortex: A PET-
percent signal change for faces as for objects (each compared to fixation.) To RCBF study of selective attention to faces and locations. J. Neurosci. 14, 63366353
(1994).
eliminate the influence of less face-selective voxels, we defined the center 6. Kanwisher, N., McDermott, J. & Chun, M. M. The fusiform face area: A module in
of the right FFA using criteria more stringent than in any published study. human extrastriate cortex specialized for face perception. J. Neurosci. 17,
The only voxels selected were found within the cluster of contiguous voxels 43024311 (1997).
selected using the less stringent criterion, showed twofold greater percent- 7. McCarthy, G., Puce, A., Gore, J. C. & Allison, T. Face-specific processing in the
age signal change for faces as objects (compared with fixation) and, in each human fusiform gyrus. J. Cogn. Neurosci. 9, 605610 (1997).
8. Puce, A., Allison, T, Asgari, M, Gore, J. C. & McCarthy, G. Face-sensitive regions in
subject, did not have less than half the percentage signal change for faces of extrastriate cortex studied by functional MRI. Neurophysiology 74, 11921199 (1996).
the voxel showing the maximum signal change for faces. 9. Sergent, J., Otha, S. & MacDonald, B. Functional neuroanatomy of face and object
processing. Brain, 115, 1536 (1992).
fMRI imaging parameters and analyses. Most (16) subjects were scanned 10. Kanwisher, N., Stanley, D. & Harris, A. The fusiform face area is selective for faces
at the Yale School of Medicine on a 1.5 T GE Signa scanner equipped with not animals. Neuroreport 10, 183187 (1999).
11. Sergent, J. & Signoret, J. L. Varieties of functional deficits in prosopagnosia. Cereb.
resonant gradients (Advanced NMR, Wilmington, Massachusetts) using Cortex 2, 375388 (1992).
echo-planar imaging (gradient echo single shot sequence, 168 images per 12. McNeil, J. E. & Warrington, E. K. Prosopagnosia: A face-specific disorder. Q. J. Exp.
slice, FOV = 40 20 cm, matrix = 128 64, NEX = 1, TR = 2000 ms, Psychol. A 46, 110 (1993).
2000 Nature America Inc. http://neurosci.nature.com

TE = 60 ms, flip angle = 60 ms). Six contiguous 7-mm-thick axial-oblique 13. Moscovitch, M., Winocur, G. & Behrmann, M. What is special in face recognition?
Nineteen experiments on a person with visual object agnosia and dyslexia but
slices aligned along the longitudinal extent of the fusiform gyrus covered normal face recognition. J. Cogn. Neurosci. 9, 555604 (1997).
most of the temporal lobe. Some subjects (two car experts and one bird 14. Assal, G., Favre, C. & Anderes, J. P. Non-reconnaissance danimaux familiers chez
expert) were scanned using coronal-oblique slices. Three more subjects un paysan. Rev. Neurol. 140, 580584 (1984).
(two car experts and 1 bird expert, I. G. et al., Soc. Neurosci. Abstr. 25, 15. Ishai, A, Ungerleider, L. G., Martin, A., Schouten, J. L. & Haxby, J. Distributed
212.9, 1999) were scanned using coronal slices on the 3 T GE scanner at representation of objects in the human ventral visual pathway. Proc. Natl. Acad. Sci.
USA 96, 93799384 (1999).
the MGH-NMR Center in Charlestown, Massachusetts. In this case, a 16. Tanaka, K. Inferotemporal cortex and object vision. Annu. Rev. Neurosci. 19,
custom bilateral surface coil was used to collect 168 images per slice in 12 109139 (1996).
near-coronal slices, 6 mm-thick. The imaging parameters were TR = 2000 17. Bornstein, B. in Problems of Dynamic Neurology (ed. Halpern, L.) 283318
ms, TE = 70 ms, flip angle = 90, 180 degrees and offset = 25 ms. (Hadassah Medical Organization, Jerusalem, 1963).
18. Lhermitte, J., Chain, F, Escouroole, R. Ducarne, B & Pillon, B. tude anatomo-
Before statistical analysis, images from the 1.5 T scanner were motion clinique dun cas de prosopagnosie. Rev. Neurol. (Paris) 126, 329346 (1972).
corrected for three translation directions and the three possible rotations 19. Damasio, A. R. & Damasio, H. & Van Hoesen, G. W. Prosopagnosia: anatomic
using SPM-96 software (Wellcome Department of Cognitive Neurolo- basis and behavioral mechanisms. Neurology 32, 331341(1982).
gy, London, UK). On the 3 T scanner, a bite bar was used to minimize 20. Dixon, M. J., Bub, D. N. & Arguin, M. Semantic and visual determinants of face
head motion. Maps of t-values and percent signal change, both correct- recognition in a prosopagnosic patient. J. Cogn. Neurosci. 10, 362376 (1998).
21. Riddoch, J. M. & Humphreys, G. W. Visual processing in optic aphasia: A case of
ed for a linear drift in the signal38, were created. Maps were spatially semantic access agnosia. Cognit. Neuropsychol. 4, 131186 (1987).
smoothed using a Gaussian filter with a full-width half-maximum value 22. Gauthier, I., Behrmann, M. & Tarr, M. J. Can face recognition really be dissociated
of two voxels, except for analyses in the center of the right FFA, where from object recognition? J. Cogn. Neurosci. 11, 349370 (1999).
regions of interests were very small and no smoothing was performed. 23. Yin, R. K. Looking at upside-down faces. J. Exp. Psychol. 81, 141145 (1969).
24. Tanaka, J. W. & Farah, M. J. Parts and wholes in face recognition. Q. J. Exp. Psychol.
In group composite maps, the percent signal change relative to fixation
A 46, 225245 (1993).
baseline for both birds and cars was multiplied with contrast weights for 25. Farah, M. J., Wilson, K. D., Drain, H. M. & Tanaka, J. W. The inverted face inversion
each subject (1 and 1 for bird experts and 1 and 1 for car experts). Under effect in prosopagnosia: Evidene for mandatory, face-specific perceptual
the null hypothesis of no expertise effect, the expected value for this contrast mechanisms. Neuropsychologia 33, 661674 (1995).
was equal to zero. We used a randomization test to asses the statistical sig- 26. Diamond, R. & Carey, S. Why faces are and are not special: An effect of expertise. J.
Exp. Psychol. Gen. 115, 107117 (1986).
nificance of percent signal changes. A population distribution for each voxel 27. Gauthier, I. & Tarr, M. J. Becoming a Greeble expert: Exploring the face
was generated by calculating randomized mean values (1000 times) of the recognition mechanisms. Vision Res. 37, 16731682 (1997).
contrast in which randomly chosen subsets of half the subjects got reversed 28. Gauthier, I., Williams, P., Tarr, M. J. & Tanaka, J. W. Training Greeble experts: A
weights. The observed contrast, calculated without sign reversal, was assigned framework for studying expert object recognition processes. Vision Res. 38,
a p value or proportion of times that the observed contrast was more extreme 24012428 (1998).
29. deGelder, B., Bachoud-Lvi, A.-C. & Degos, J.-D. Inversion superiority in visual
than the randomized contrast). To show the anatomy clearly, the p values agnosia may be common to a variety of orientation-polarised objects besides faces.
were overlaid on the normalized anatomical images for a single subject Vision Res. 38, 28552861 (1998).
(threshold at p < 0.01; Fig. 6). 30. Gauthier, I., Anderson, A. W., Tarr, M. J., Skudlarski, P. & Gore, J. C. Levels of
categorization in visual object recognition studied with functional MRI. Curr. Biol.
7, 645651 (1997).
Note: Additional analysis can be found on the Nature Neuroscience web site 31. Halgren E., Dale, A. M., Sereno, M. I., Tootell, R. B., Marinkovic, K. & Rosen,
(http://neurosci.nature.com/web_specials/). B. R. Location of human face-selective cortex with respect to retinotopic areas.
Hum. Brain Mapp. 7, 2937 (1999).
32. Rolls, E. T. & Baylis, G. C. Size and contrast have only small effects on the response
ACKNOWLEDGEMENTS to faces of neurons in the cortex of the superior temporal sulcus of the monkey.
We thank Nancy Kanwisher and Ren Marois for discussions and Jill Moylan, Exp. Brain Res. 65, 3848 (1986).
Terry Hickey and Hedy Sarofin for technical assistance. This work was supported 33. Rosnow, R. L. & Rosenthal, R. Some things you learn arent so: Cohens paradox,
Aschs Paradigm, and the interpretation of interaction. Psychol. Sci. 6, 39 (1995).
by NINDS grant NS33332 to J.C.G. and NIMH grant 56037 to N. Kanwisher. 34. Epstein, R., Harris, A., Stanley, D. & Kanwisher, N. The parahippocampal place
I.G. was supported by NSERC. area: Recognition, navigation, or encoding? Neuron 23, 115125 (1999).
35. Puce, A., Allison, T, Asgari, M, Gore, J. C. & McCarthy, G. Differential sensitivity of
human visual cortex to faces, letterstrings, and textures: A functional magnetic
RECEIVED 11 AUGUST; ACCEPTED 7 DECEMBER 1999 resonance imaging study, J. Neurosci. 16, 52055215 (1996).
36. Wojciulik, E., Kanwisher, N. & Driver, J. Modulation of activity in the fusiform face
area by covert attention: an fMRI study. J. Neurophysiol. 79, 15741578 (1998).
1. Archambault, A., ODonnell, C. & Schyns, P. G. Blind to object changes: When 37. Tranel, D., Logan, C. G., Frank, R. J. & Damasio, A. R. Explaining category-related
learning the same object at different levels of categorization modifies its perception. effects in the retrieval of conceptual and lexical knowledge for concrete entities:
Psychol. Sci. 10, 249255 (1999). operationalization and analysis of factors. Neuropsychologia 35, 13291339 (1997).
2. Allison, T. et al. Face recognition in human extrastriate cortex. J. Neurophysiol. 71, 38. Skudlarski, P., Constable, R. T. & Gore, J. C. ROC analysis of statistical methods
821825 (1994). used in functional MRI: Individual subjects. Neuroimage 9, 311329 (1999).

nature neuroscience volume 3 no 2 february 2000 197

You might also like