Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

PERSPECTIVES

function6. These discoveries provided crucial


TIMELINE
tools for subsequent experimentation. Bell
also expressed the idea that sensory nerves
Ideas about pain, a historical view were specialized in their function, that is,
particularly adapted to detect and carry
information about a given stimulus mode.
Edward R. Perl Later, the concept of sensory nerve specifi-
city was developed by Müller (1840), whose
Abstract | The expression ‘painful’ can be used to describe both an embarrassing
postulates proved both seminally important
moment and a cut on the finger. An explanation for this dichotomy can be found in subsequent thinking and a focus for
in the convoluted history of ideas about pain. Whether pain is an independent criticism1,2,7. Müller’s proposals were given
sensation and the product of dedicated neural mechanisms continues to be a topic impact by the contemporary description
of debate. This overview concentrates on the issue of specificity together with of the electrochemical nature of the
other notable information regarding pain that has emerged since 1800. nerve impulse by DuBois-Reymond8. If
the impulses of different nerve fibres were
similar, peripheral and central endings
The word ‘pain’ and its synonyms Much attention was given to nervous of afferent fibres must provide part of the
commonly refer to conscious experiences system anatomy after the Middle Ages; information about the nature of the stimulus
associated with bodily injury or disease, however, progress in thinking regarding pain to the brain.
but are also used to describe discomfort awaited developments in understanding of Mid-nineteenth century studies of
related to other unpleasant feelings. These the natural world and scientific tools. For spinal cord pathways added important
different circumstances are the bases of a example, until the beginning of the nine- insights. Schiff (1858), once Magendie’s
long-standing uncertainty regarding how teenth century, ideas about sensation were student, showed that particular lesions of
to think about pain and its mechanisms. mired in the belief that the senses depend on the spinal cord resulted in separate and
This article aims to provide a historical the transport of an agent from the outside independent loss of tactile and pain-related
perspective on changing views over the world to the heart or brain. That concept lost reactions9. This led Schiff to propose pain
past two centuries. favour in the eighteenth century due in part to be an independent sensation, mirroring
There is a legacy from distant times. to changing insight into the physical world conclusions reached by Avicenna a millen-
Aristotle (384–322 bc) considered the and proposals by Newton (1642–1727) and nium earlier. Schiff ’s experimental findings
heart to be the seat of feelings. Taking Hartley (1705–1757) that neuronal messages were confirmed and expanded by Brown-
cognizance of pain’s usual importance for were vibrations of substance in nerves2. Séquard (1860; 1868), who documented
disposition, he argued it to be an emotion. This article highlights the sequence of the loss of pain sensibility contralateral
Not all ancient Greek philosophers agreed; ideas and observations since 1800 that have and distal to a transverse hemisection of
however, Aristotle’s influence on philo- contributed to concepts about the nature of the spinal cord10,11, and Gowers (1878),
sophical thought endowed his views with pain and its neural mechanisms (TIMELINE). a prominent English neurologist, who
an enduring impact. Centuries later, Galen Although it is a focused history, it also pro- reported similar effects following a lesion
(ad 130–201), a leading physician–surgeon vides insight into progress in understanding in a human2,12. By the 1880s there were
of Alexandria, used experimental studies the function of the mammalian nervous several lines of support for a spinal path-
along with earlier observations to disagree. system. way essential for conducting information
Galen recognized the brain as the organ on painful stimuli.
of feeling and placed pain into the sphere of Nineteenth century issues However, not all observers accepted
sensation. Avicenna (ad 980–1037), Afferent pathways for somatic senses. How the notion that pain was an independent
a renowned Muslim philosopher and different information from the body is trans- sensation. The German neurologist Erb
physician, noted that, in disease, pain can mitted to and from the spinal cord and brain (1874), noting that intense sensations are
dissociate from touch or temperature recog- was seriously addressed in the nineteenth usually disagreeable and involve strong
nition, and proposed pain to be an inde- century. In 1811, Bell, a Scottish physician stimuli, proposed pain to be the outcome
pendent sensation1–3. Despite much work and anatomist, proposed that the dorsal and of vigorous activation of nervous pathways
and thought since these ideas were aired, ventral spinal roots differ in function, with normally concerned with other sensory
fundamental issues about pain remain the ventral roots being responsible for experiences13. Although it had been hinted
unresolved. Notably, these include whether control of muscle contraction5. However, at earlier, Erb’s proposal gave birth to an
pain results from the activity of a dedicated Bell was vague about the role of the dorsal explicit theory of pain, which presumed
neural apparatus or is the product of less roots and Magendie (1822) is generally that intensity of stimulus and response were
specific processes4. credited with demonstrating their sensory the main factors (FIG. 1b).

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | JANUARY 2007 | 71


© 2007 Nature Publishing Group
PERSPECTIVES

Timeline | Important discoveries and concepts for mechanisms related to pain over the past two centuries

(1924–1928)
Schiff describes von Frey finds Erlanger, Gasser and
Bell proposes differential effects of circumstantial Bishop describe
difference in spinal cord lesions on correlations Spiller and Martin compound action
function of reactions to peripheral Blix and Goldscheider between sensory use ventrolateral potential of
spinal dorsal Müller postulates and non-painful stimuli Erb proposes demonstrate mosaic skin spots and spinal chordotomy peripheral nerve and
and ventral specific function of and proposes pain to be intensive theory of of skin sensation and structure of for the relief of pain relationship to fibre
roots5 afferent neurons7 a specific sense9 pain13 pain spots14,15 nerve endings18 in humans25 diameter30,135–137

1811 1822 1840 1848 1858 1860 1874 1878 1884 1890 1896 1906 1912 1915 1924 1926

Magendie shows (1848–1849) Brown-Séquard Gowers reports Edinger identifies the Sherrington Ranson argues Adrian and
that spinal dorsal DuBois-Reymond demonstrates dissociation of pain crossed afferent tract proposes definition small diameter Zotterman
roots transmit identifies the nerve dissociation of pain from touch after a in the spinal cord of pain-causing primary afferent record action
sensory messages6 action potential8 from touch following spinal cord lesion in (spinothalamic)22 stimuli as tissue- fibres are potentials from
experimental spinal a human12 damaging (noxious)20 protopathic and individual
lesions10,11 evoke pain29 afferent fibres37
(1906–1911) Dejerine and Roussy, and
Head and Holmes postulate a role for
the thalamus in pain26,27

So, near the end of the nineteenth cen- correlation between skin spot and nerve This pathway was linked to pain by the
tury, three contrasting concepts about the terminal histology were particularly in effects of experimental lesions in both
nature and mechanisms of pain prominently question. animals and clinical cases2. Spiller and
and vigorously competed: an emotion Martin (1912) provided a ‘proof of concept’
(from ancient philosophy, supported by Nociception and neural circuits. One by reporting a case in which transection of
philosophers and some psychologists); a objection to considering pain as a specific the ventrolateral spinal cord was used to
specific sensation with its own sense organs sense was its evocation by different types of treat persisting, intractable pain from the
and pathways (formulated by Avicenna and stimuli (mechanical, thermal and chemical), opposite side of the body25. Observations on
Schiff, supported by physiologists and physi- which differed from accepted sensations. clinical syndromes led Dejerine and Roussy
cians); and intense activation of afferent sys- Sherrington (1906), noting that pain (1906) and Head and Holmes (1911) to
tems that serve other sensations (proposed commonly originated from tissue injury, implicate the thalamus of the rostral brain
by Erb, supported by many psychologists suggested labelling stimuli capable of tissue in the production of pain26,27.
and some physicians)1. damage as ‘noxious’ regardless of physical Müller’s proposal in 1840 of specific
New considerations and information character20. He proposed the signalling of functions of individual sensory nerves7
came to light at this point. In 1884, Blix14 noxious events (nociception) to be the func- implies that different somatic sensations
and Goldscheider15 noted cutaneous sensa- tion of sense organs responsible for pain. are served by separate and distinctive nerve
tion to be spatially discontinuous. Separate This concept endowed pain with coherent fibres. Consistent with this, nineteenth cen-
spot-like areas of the skin yielded different and definable peripheral stimuli. tury anatomists had noted that dorsal roots,
sensory experiences (pressure, cold, Recognition in the 1890s that the CNS is on joining the spinal cord, separate into a
warmth, pain) when stimulated mechani- composed of discrete cells, rather than being a medial division containing large-diameter
cally with small probes. The observations syncytium was another important conceptual fibres and a lateral division composed of
of von Frey (1896/1897) strongly supported step21. The concept of synapses as functional thin fibres23. Observations on the regenera-
this concept and went further, comparing connections between cells also supported tion of transected nerves and the return of
the relative numbers of particular types Sherrington’s seminal idea that, in addition to sensation led Head and colleagues in 1905
of spot to the numbers of histologically excitation, inhibition of excitability in neural to postulate two different classes of somatic
defined neural structures in the same circuits is essential for integrative function- sensory innervation, epicritic (discrimina-
regions. From this he deduced relationships ing20. So, nervous pathways could be envi- tive) and protopathic (crude)28. Diffuse,
between the types of spot and structurally sioned to consist of neurons with functional aching pain was of the protopathic genre.
defined neural endings16–19. This analysis connections that could be altered. A decade later, Ranson proposed that fine,
did much to convince physicians and unmyelinated fibres of the lateral dorsal root
physiologists that pain is an independent 1900–1965: new approaches division are protopathic, and in part repre-
sense. Nonetheless, sceptics remained: Anatomists (Edinger, 1890, 1892; Bechterew, sent an afferent limb for pain29.
Goldscheider swung from supporting the 1900) provided convincing evidence that
existence of separate pain spots to denying a long ascending pathway originated from Function of individual neurons: electrophys-
them in favour of a version of the intensive spinal neurons whose axons crossed the iology. Early in the twentieth century, record-
concept, famously battling von Frey in the midline and ascended in the lateral white ing of electrical signals from nerves added
literature1. von Frey’s deductions on the matter to end in the rostral brainstem22–24. a new dimension. Using the cathode-ray

72 | JANUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro


© 2007 Nature Publishing Group
PERSPECTIVES

(1973–1977) Price (1981–1983)


and Mayer describe Microneurographic
Christensen and Perl stimulation in Caterina et al. identify
multireceptive (wide
describe selective conscious humans capsaicin receptor that
dynamic range)
Zotterman nociceptive neurons in shows that endows responsive features
neurons in
Nafe suggests reports that (1948–1955) spinal marginal zone84 excitation of to polymodal nociceptors61
spinothalamic
pattern theory some thin Oxford anatomists’ pathway92 nociceptors, but
for definition of afferent fibres challenge to not of low- Rainville et al. use positron emission
stimulus are activated neuronal specificity Burgess and Perl threshold tomography and functional MRI to
features by by gentle in the cutaneous document myelinated Pert and Snyder identify mechanoreceptors, show differential anterior cingulate
nerve activity38 stimuli40 sense42,43 fibre nociceptors57 opiate receptors131 evokes pain80,81 cortex activation by painful stimuli124

1929 1933 1939 1942 1948 1965 1967 1969 1970 1972 1973 1975 1981 1983 1997 2003

Heinbecker, Bishop (1942–1952) Lewis, Melzack and Bessou and Perl Identification of Hökfelt et al. Woolf demonstrates Craig proposes pain
and O’Leary show and Hardy, Woolf and Wall propose describe C-fibre spinothalamic show expression central sensitization as a homeostatic
that pain in humans Goodell describe gate control polymodal neurons by of peptides in spinal nociceptive emotion4
is related to primary and secondary theory of pain54 nociceptors and antidromic and (substance P, pathways97
activation of thin hyperalgesia71,72 nociceptor retrograde calcitonin gene-
fibres31 sensitization59 tracing101–103 related peptide)
in small primary
afferents63

oscilloscope, Erlanger, Gasser and Bishop30 went on to postulate that the mode of stimu- undifferentiated afferent neurons signalled
showed that a brief electrical stimulus to a lation is signalled by the composite of activ- the nature of stimulation. Wall, who later
peripheral nerve evokes a series of conducted ity in a population of afferent nerve fibres, had an important part in developing con-
electrical waves (FIG. 2). These studies on thereby creating another hypothesis for the cepts regarding pain, was a student at Oxford
the compound action potential provided an coding of nerve activity leading to bodily when these views were generated.
opportunity to correlate afferent fibre activity sensations — the pattern theory (FIG. 1c). By the mid-twentieth century, electronic
with sensory experience. Between 1930 and 1950, it was established equipment capable of amplifying and display-
Studies by Bishop, Gasser and their that most rapidly conducting, large-diameter ing small electrical signals had markedly
collaborators showed that selectively exciting afferent fibres could be excited by a particular improved, facilitating electrophysiological
or blocking activity in fibres contributing form of mechanical stimulation of the skin recording from thinly myelinated Aδ fibres
to components of compound potentials or subcutaneous tissue. However, recording and unmyelinated primary afferent C-fibres.
implicated thinly myelinated (Aδ-) and activity from the finest afferent fibres proved In the 1950s and early 1960s, several investi-
unmyelinated (C-) fibres in pain and aver- technically challenging. Notably, in 1936, gators (Tasaki’s group45, Paintal46, Bessou
sive reactions31–35. Furthermore, as Lewis and Zotterman concluded that the slowest fibres and Laporte47, and Hunt and McIntyre48,49)
Pochin later reported, a single brief noxious (C category) in the lingual nerve signal ‘pain’ reported that some slowly conducting myeli-
cutaneous stimulation initiates a double pain because they were excited only by strong nated afferent fibres required strong (noxious)
experience, with the time between the two stimuli39. Three years later, however, he stimuli for activation. Iggo’s notable papers
pains being proportional to the conduction reported that some C-fibres innervating the in 1959 and 1960 showed that certain single
distance34,36. The latter finding indicates that cat skin were activated by light tactile stimu- unmyelinated fibres from the skin give selec-
afferent fibres of differing conduction veloci- lation40. The paucity of these observations tive responses to noxious stimuli50,51; however,
ties are involved in pain of peripheral origin. raised doubts about the specificity of periph- the Oxford challenge to specificity could have
In the mid-1920s, natural stimulation eral afferent neurons conveying activity that blunted their impact, even though there were
(for example, brushing/stroking of skin, produced pain. If pain receptors existed, in comparable observations by Iriuchijima and
stretching muscles, temperature changes and 1940 their features remained obscure. Zotterman52. The extensive survey of myeli-
so on) of peripheral tissue was used to evoke nated afferent units serving mammalian skin
discharges in individual afferent fibres. The The Oxford challenge to sensory nerve by Hunt and McIntyre noted only a handful
early galvanometer records published from specificity. Between the late 1940s and to have high thresholds to mechanical stimu-
Adrian’s Cambridge laboratory showed mid-1950s, members of Oxford University’s lation, a point that later was to figure in an
a series of irregular potential deflections Department of Anatomy voiced strong excep- analysis arguing against specific sense organs
evoked in fine nerve filaments by various tion to the concept of specific signalling by for pain. Although these studies described
distinctive stimuli. These records were cutaneous afferent fibres. This was based on afferent neurons responsive only to strong
interpreted to represent selective responses an absence of correlation between the modal- (noxious) stimuli, and therefore could be
by different nerve fibres to particular forms ity of sensation evocable from the human taken to support the specificity theory, their
of stimulation37. Nafe (1929), an American pinna or cornea and histological analyses small numbers, the lack of consistent descrip-
psychologist, challenged this view, proposing of the innervation of these tissues41–44. They, tions and the presence of units responsive to
instead that the pattern of discharge changes like Nafe, proposed that a pattern of nerve weak stimuli among the thin-fibre population
with different forms of stimulation38. Nafe impulses in otherwise functionally weakened the case.

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | JANUARY 2007 | 73


© 2007 Nature Publishing Group
PERSPECTIVES

A review in 1962 on cutaneous sensation Their argument emphasized the ambivalence postulating the operation of a neural gate in
by Melzack and Wall accepted the existence of data correlating given cutaneous sensa- the dorsal horn of the spinal cord to control
of differences in the responsiveness of affer- tions with histologically definable primary activation of ascending projections54 (FIG. 1d).
ent fibres, but concluded that spatial and afferent terminations. Three years later, they Absence of specific pain receptors and a lack
temporal patterns of nerve activity form the elaborated a pattern-based theory (the gate of dedicated central pathways are core to this
basis of sensory perceptions from the skin53. theory) about mechanisms underlying pain, proposal. The gate theory was proposed in
opposition to the specific-sense concept.
a Specificity theory b Intensity theory It was emphasized that despite many years
of study, few primary afferent fibres or
CNS cells were reported to be selectively
activated by pain-causing stimuli. This
Impulses

Impulses
was buttressed by reports of the absence of
complaints of pain by soldiers at the time
of grievous injury and the occurrence of
pain in CNS disorders, such as thalamic
syndrome, in which noxious stimuli were
Innocuous Noxious Innocuous Noxious
not involved. The gate theory presumed
Intensity Intensity
primary afferent fibres to have differences
DRG Low threshold in adaptation to maintained stimuli and
Skin nociceptors Dorsal horn Skin DRG neurons WDR dorsal
nociceptive horn projection to represent a continuous spectrum of
Noxious neurons Noxious neurons responsiveness. It postulated that afferent
stimulus stimulus
signals from thick and thin fibres interact
Innocuous in the substantia gelatinosa of the dorsal
stimulus
horn of the spinal cord. Applying the estab-
c Pattern theory d Gate control theory lished principle of control of spinal cord
mechanisms by supraspinal centres20,55,56,
Cell 1
their theory included modulation of the
Central
Impulses

control
gate by higher centres. Despite questions
Cell 2 about the validity of the underlying
assumptions, the theory was received with
Cell 3 excitement and broadly accepted, particu-
Gate control system larly because it offered possible explana-
Innocuous Noxious A-fibres – tions for aberrant pain after lesions of the
+ +
Intensity Action
SG T nervous system.
– system
DRG +
neurons –
Skin Documentation of nociceptors since 1965
Cell 1 Dorsal horn C-fibres
Noxious The presumption that afferent fibres
neurons
stimulus Cell 2
selectively responsive to tissue injury
Innocuous (nociceptors) do not exist is crucial to the
stimulus Cell 3 intensive, pattern, and gate theories. This
postulate was shown to be untenable in 1967
Figure 1 | Theories of pain. Diagrams depicting typical assumptions about relationships between by Burgess and Perl, who documented a
stimuli and primary afferent signalling in theories about pain. a | According to the specificity theory, substantial proportion of slowly conducting
specialized sense organs (nociceptors) have thresholds at or near noxious levels, increasing activity myelinated fibres in cat cutaneous nerves to
with stronger noxious stimuli. These special peripheral afferent neurons have selective connections have distinct nociceptive characteristics57.
to particular spinal and brainstem projection neurons. b | The intensity theory suggests that peripheral
Such afferent fibres are selectively respon-
sense organs are not differentiated into low- and high-threshold types. It proposes that afferent fibres
transduce innocuous stimuli (for example, skin pressure) by generating a certain level of activity, sive to strong mechanical stimuli, grading
whereas noxious stimuli are signalled by a greater level of discharge. The intensity-coded primary activity proportional to noxious stimulus
afferent fibres, in turn, activate projection neurons with a wide dynamic range (WDR). Weak activation intensities. Similar nociceptive afferent fibres
of WDR projections indicates innocuous stimuli; strong activation indicates painful (noxious) events. are present in primates58. A subsequent
c | The pattern theory proposes that somatic sense organs have an extensive range of responsiveness. study by Bessou and Perl pointed out that a
Individual afferent neurons respond to stimuli with differing relationships to intensity. The mode and large proportion of cat cutaneous C-fibres
locus of stimulation are indicated by the composite pattern of activity in the population of fibres from express coherent nociceptive features, being
a particular body region. Central projection neurons code the nature and place of stimulation by the effectively excited by noxious heat, strong
pattern and distribution of their discharges. d | According to gate control theory, the spectrum of mechanical stimuli, and acid59. These experi-
primary afferent neurons has a range of thresholds, specialized nociceptors and dedicated central
ments differed from prior work by utilizing
pathways do not exist, and large-diameter primary afferent fibres (A-fibres) adapt more quickly to
maintained stimuli than thin ones (C-fibres). A presynaptic gate in the substantia gelatinosa (SG) of the recording methods that sampled peripheral
spinal dorsal horn between primary afferent and projection neurons is controlled by the balance of nerve fibres more randomly.
activity between the A-fibres and the C-fibres. When the C-fibre input outweighs that of the A-fibres, A substantial body of evidence has since
the gate opens, permitting activation of projection neurons. CNS mechanisms (descending control) accumulated indicating nociceptors to be
are postulated to modulate the gate. DRG, dorsal root ganglion; T, transmission neurons. distinctive afferent units rather than the

74 | JANUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro


© 2007 Nature Publishing Group
PERSPECTIVES

limits of a population with a continuum of


features. Nociceptors discriminate reliably Aα,Aβ
between noxious and innocuous stimulation, 6
100
a capability that is missing in low-threshold
mechanoreceptive or thermoreceptive affer- 80

Velocity (ms–1)
ent neurons58,60. Nociceptors differ from 5 60
low-threshold afferent fibres not only by 40
having elevated thresholds for all stimuli 20
ordinarily affecting a tissue, but also in their 4
0
membrane constituents61 and membrane

Amplitude (mV)
0 2 4 6 8 10 12 14
properties, which include action potential Diameter (μm)
shape62 and cytochemistry63–65. Furthermore, 3
they have distinctive termination patterns in
the spinal cord66,67. A number of tissues have
been found to be innervated by more than 2
one category of nociceptor with different
responsiveness to various noxious stimuli65. Aδ
0 0.4 0.8 1.2 1.6 2.0 2.4 2.8 3.2
Therefore, at the beginning of the twenty-first 1 Time (ms)
century, nociceptors are widely accepted as
separate classes of primary sensory neurons, C
with elevated thresholds to all forms of 0
natural stimuli and selectivity in their
response to different modes of stimulation.
0 10 20 30 40 50 60 70 80 90
Time (ms)
Sensitization of nociceptors. It has become
Figure 2 | The compound action potential. This composite figure depicts the compound action
increasingly clear that signals transmitted
potential of a mammalian cutaneous nerve. The outer graph is a scale drawing of the compound
by nociceptive afferents to the CNS are action potential of a saphenous nerve for a conduction distance of 37 mm. The wave peaks are labelled
influenced by past events. First described alphabetically in order of latency30. Gasser’s and Erlanger’s elegant analyses in 1927 established the
in frogs by Echlin and Propper (1936) and first peak (A) and its subdivisions (Aα, Aβ, Aδ) to be the summed electrical activity of myelinated
subsequently noted in cats by Witt and fibres135. A much delayed (slowly conducting) C deflection was later found to represent the summed
Griffin (1962), once they are activated high- action potentials (nerve impulses) of unmyelinated fibres136. The speed at which action potentials
threshold afferent fibres exhibit a notable are conducted was postulated to vary as a positive function of each fibre’s cross-sectional diameter.
propensity to increase their responsive- Fibre conduction velocity is distributed around certain modal values, giving rise to the various deflec-
ness to subsequent stimuli65,68–70 (FIG. 3). tions in the compound potential137. The inset shows the compound action potential of myelinated
Enhancement of nociceptor responses as primary afferent fibres (A-fibres) from a saphenous nerve on a faster time base to depict the summation
of action potentials of different fibres. Triangles, scaled in amplitude and duration to reflect dimensions
a consequence of tissue injury might be
of impulses recorded from fibres of particular conduction velocities, were used to represent each
at least partially the result of modification fibre of the nerve identified in histological cross-section. The triangles are positioned on the horizon-
of the extracellular milieu by intracellular tal axis at the latency calculated from the conduction velocity derived from the fibre’s measured
contents issuing from injured cells (such as diameter. Top right graph shows the relationship between fibre diameter (abscissa) and conduction
protons, potassium ions or ATP). Following velocity (ordinate) by the line as predicted by assumption and by dots as measured. Reproduction of
tissue damage, the complex process of the compound potential by summing of the triangles closely fit the recorded potential, consistent with
inflammation adds another set of agents validity of the summation and conduction velocity postulates. Modified from REFS 138,139.
(for example, serotonin, bradykinin, prosta-
glandins, growth factors62 and cytokines)
and circumstances that alter the responsive- markedly decreases and the proportion of (impulses) in nociceptive afferent fibres
ness of peripheral nociceptor terminals. mechanically excitable fibres increases73–75. and behavioural measures59,76,77. Whereas
Sensitization of afferent fibres by processes The colourful names ‘sleeping’ or ‘silent’ nociceptor responses in experimental
operating on their peripheral terminal is nociceptors have been given to afferent animals correlate well with descriptions
presumed to be partially responsible for fibres that are unresponsive under normal of human pain evoked by the same
increased pain (primary hyperalgesia) in conditions. Presumably, they represent a stimuli, such comparisons depend on an
injured tissue64,65,70–72. category of sensory nerve fibres lacking assumption of similarity between species.
mechanical responsiveness until exposed to Hagbarth and colleagues, particularly
Silent nociceptors. Systematic surveys of an inflammatory environment. The recruit- Torebjörk, established the technique of
slowly-conducting afferent fibres in normal ment of silent joint nociceptors to an active microneurography — the sampling of elec-
individuals describe some to be unresponsive state is a possible important contributor to trical activity from individual nerve fibres
to any intensity of mechanical or thermal arthritic pain. in conscious human subjects — making
stimulation. This is particularly evident in direct comparisons in a given person possi-
the innervation of musculoskeletal tissue. Relationship of nociceptors to sensation. ble77–79. Torebjörk described close parallels
As Schaible and Schmidt pointed out, Extensive information connecting activ- between the activity of human cutaneous
when inflammation is present, the number ity in nociceptors to sensation has come nociceptors and the individual’s reports of
of inexcitable nerve fibres from joints from correlations between evoked activity painful sensations77.

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | JANUARY 2007 | 75


© 2007 Nature Publishing Group
PERSPECTIVES

Using the recording microelectrode in stimulation. Christensen and Perl (1970) limb or the body, and they respond much
human microneurography to electrically showed that some neurons in spinal lamina I as predicted by the intensive theory (FIG. 1).
stimulate nerve fibres at the recording locus (marginal layer of the spinal dorsal horn) The presence of both selectively nociceptive
was suggested by Konietzny et al. and by are selectively activated by stimuli exciting neurons and the multimodal, WDR type of
Torebjörk and Ochoa; it provides compelling, nociceptors or thermoreceptors (afferent response in the mammalian spinal dorsal
albeit controversial, indications that activity neurons specifically activated by innocuous horn has been confirmed and expanded by
in nociceptors, but not low threshold mech- warming or cooling)84. Craig has presented a number of investigations4,86.
anoreceptors, evokes painful experiences80,81. pivotal evidence that many lamina I neurons Both selective and WDR neurons
The interpretation of such microstimulation projecting to higher centres are selectively participate in nociceptive and pain-related
studies presumes that weak electrical currents responsive to noxious and/or thermal mechanisms. The differences in their coding
applied through the microelectrode excite the stimuli. The lamina I region has been shown of information from the periphery suggest
closest fibres, those from which discharges are to receive direct input from myelinated- and that they serve different functions in the
recorded. Objections to that interpretation by unmyelinated-fibre nociceptors66,67,84–86 and organization of sensory and reflex reactions.
Wall and McMahon focus on the large size of also to contain neurons with focused respon- Selective responsiveness of lamina I neurons
the metal microelectrode tip compared to the siveness to pruritic stimuli4. Importantly, provides a basis for differentiating noxious
diameter of the fine nerve fibres82; however, both human and animal studies indicate that and innocuous events, a distinction made by
the same size discrepancy exists for the lamina I neurons contribute to the classical the receptive characteristics of the primary
recording of single-fibre traffic with micro- spinothalamic pathway87–89. afferent neurons. This specificity is presum-
neurography electrodes83. Furthermore, the A different and widely studied type of ably carried forward by the focused respon-
microneurography stimulation descriptions spinal neuron features a multimodal input siveness of lamina I neurons. The WDR type
relate a remarkable correspondence between from primary afferent fibres and is largely of response might concern mechanisms
receptive fields of the recorded afferent fibres found deeper in the dorsal horn (laminae other than those signalling the nature and
and the projected locus of the sensation V–VI). Such multimodal neurons evidence location of stimuli, although some proposals
evoked by intraneural microstimulation80,81,83. weak activation by innocuous mechanical presume that the convergent WDR neurons
stimulation and more intense excitation by could provide the information necessary for
Late 20th century: CNS arrangements noxious mechanical or thermal stimuli; they both modality and location designation4,93.
Documentation of the characteristics of are often referred to as wide dynamic range It is possible that the large receptive fields of
nociceptors provided tools to refine the (WDR) neurons90–92. Their receptive fields multimodal projection neurons function in
search for CNS activity related to noxious are extensive, involving large regions of a mechanisms that set the responsiveness of
higher centres94.
There is now convincing evidence that
a b projections of both selective and non-selec-
200 tive neurons with nociceptive features reach
similar levels in the forebrain86,95. Lamina
I and laminae V–VI spinal neurons send
axons to the midbrain and thalamus93,95,96.
Threshold temperature (°C)

50
150
To some extent these projections might
Number of impulses

converge. Circumstantial evidence suggests


features of WDR-type ascending projec-
100 tions to fit those capable of evoking pain
in conscious human subjects91. However,
40 spinal chordotomy in humans in which the
lateral tracts of the spinal cord are divided
50
surgically to treat otherwise uncontrollable
pain from the contralateral body25 provides a
circumstantial link between human lamina I
0 30 neurons and pain. The segmental distribu-
1 2 3 4 5 1 2 3 4 5 tion of lamina I neurons showing retrograde
Number of stimulations Number of stimulations degeneration from the lesion of the
Figure 3 | Peripheral sensitization of nociceptors. Exposure to noxious heat is associated with an contralateral spinal cord fits closely to the
increased response and a decrease in heat threshold by mammalian single unmyelinated (C-) fibre distribution of the lost pain sensitivity87,88.
nociceptors. Action potentials from C-fibres from primate polymodal nociceptors were isolated by Noxious stimulation also evokes
mechanically teasing a cutaneous nerve. A small contact thermode was used to produce a sequence enhanced pain in tissue uninjured by the
of temperature increases to stimulate the skin (stepping in equal increments from 30–53°C in ~90 s event. This remote effect has been attrib-
followed by quick cooling to the 30°C holding temperature). Identical stimulus programs were uted to central nervous mechanisms72,97,98.
repeated at 200 s intervals. a | The total number of impulses between the start of the heating program
Vigorous activity in certain nociceptive
and the start of the next heating cycle. b | The temperature threshold for evoked impulses in each run.
The symbol indicating a given fibre also applies to both panels. Note the increase in number of impulses afferents is associated with an increased
generated during a given cycle and the decrease in the threshold temperature on successive applica- synaptic activation of spinal neurons; phe-
tions of the heat stimuli. The increased response and lowering of threshold is evident in some units nomena now labelled ‘central sensitization’.
after a single stimulus cycle. Reproduced, with permission, from REF. 140 © (1977) American Central sensitization could be the product
Physiological Society. of several different processes. Presynaptic

76 | JANUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro


© 2007 Nature Publishing Group
PERSPECTIVES

terminals of many neurons, including some multireceptive characteristics, fitting the Early in the twentieth century, pain
primary afferent fibres, contain more than WDR category95. How this mix of signals was suggested to be a product of thalamic
one excitatory synaptic mediator. One and cell groups is organized to provide a mechanisms, discounting a contribution
mediator (for example, glutamate) could sensation remains unknown. by the cerebral cortex based on absence of
be released on infrequent to moderate The crossed ventrolateral pathway is selective alteration in pain perception after
activation, with another (such as a peptide not the sole ascending spinal projection to lesions or electrical stimulation26,27,115,116.
or ATP) being released when there are high carry nociceptive information; however, However, there were also reports of a persist-
levels of presynaptic activity99. Secondary in general, there is a lack of convincing ing loss of capacity to recognize painful
mediators could enhance the response evidence for important contributions from stimulation in restricted body regions after
to other agents or increase excitability of other major ascending pathways in normal localized cortical injuries in the central
postsynaptic neurons100. Alternatively, nociceptive reactions and pain86. An excep- sulcus region. These earlier observations and
a given mediator such as glutamate can tion is the relatively recent uncovering by modern imaging analyses strongly support
activate different postsynaptic excitatory Willis and colleagues of a role for the spinal the concept that the cerebral cortex has an
receptors — for example, AMPA (α-amino- dorsal columns in transmitting pain-related important role in pain processing117–120.
3-hydroxy-5-methyl-4-isoxazole propionic information from pelvic viscera to higher
acid), NMDA (N-methyl-d-aspartate) centres106–108. This dorsal spinal pathway Imaging. The last decade of the twentieth
and metabotropic receptors — again on helps to explain the clinical observation century saw an explosion of information
an activity-dependent basis. Activation that a lesion of the spinal cord in the dorsal from imaging blood flow and fluid changes
of NMDA receptors induces a persisting midline, separating the dorsal columns, can in the intact, functioning human brain
facilitation, part of a process leading to alleviate visceral pain109. using positron emission tomography (PET)
the generation of long-term potentiation Whereas certain nuclear groups of the and functional MRI (fMRI). The funda-
of synaptic effectiveness. Involvement of thalamus are a principal target of the spinal mental tenet in these studies is that blood
NMDA receptors in the central sensitization ventrolateral system, direct connections flow and fluid changes in the brain parallel
of nociceptive connections has been argued from neurons of several spinal regions to local neuronal activity. An increase in the
by Woolf and his colleagues to indicate a other higher centres including the hypotha- net metabolism of a group of neurons is
mechanism similar to memory processing lamus and parts of the amygdala have generally a consequence of excitation; how-
in the hippocampus98. been established110–112. The amygdala and ever, synchronous activation of inhibitory
hypothalamus are usually considered to linkages will leave metabolic traces as well.
Central ascending pathways. Fibres in the lat- participate in affective and homeostatic This burgeoning field of in vivo investiga-
eral and ventral spinal white matter form an functions rather than somesthesis, raising tion so far lacks convincing resolution at the
important ascending pathway for nociceptive the possibility that special pathways subserve cellular level. Nonetheless, brain imaging
and pain-related activity from the opposite nociceptive mechanisms other than sensa- spearheaded by several groups (Bushnell;
side of the body. However, much evidence tion (see below). Nociceptive signals also Derbyshire; Davies) demonstrates the
has accumulated supporting a far more reach higher brain centres via multisynaptic extent and pliability of the cerebral neural
complex arrangement than a single invariant routes by way of connections in subthalamic networks that become active after a noxious
and dedicated tract. Fibres of the crossed brainstem nuclei. stimulus and emphasizes the complexity of
ventrolateral spinal pathway were established the system. Such studies make it clear that
to reach the thalamus, giving rise to the The thalamus and cerebral cortex. The processing of information in the neural
designation ‘spinothalamic.’ Willis and his crossed ventrolateral pathway terminates systems associated with nociception and
colleagues have utilized antidromic activity in both lateral and medial thalamic pain partly occurs in several brain regions
evoked by electrical stimulation in thalamic nuclei4,95. The lateral component con- operating in parallel.
structures to uncover functional characteris- tributes terminals to several nuclei of the To a considerable degree, PET and fMRI
tics of neurons contributing to ventrolateral ventral posterior thalamus, a main soma- determinations of human brain regions acti-
tracts. Importantly, modern histochemical tosensory zone with spatial and modality vated by painful stimuli are consistent with
(anterograde and retrograde transported characteristics suggestive of a role in dis- expectations from modern morphological
markers)101,102 and physiological (antidromic) crimination64,95,96. By contrast, the medial and physiological tracing and projection
tracing86,93,103 have shown that axons in the thalamic ventrolateral projection targets studies of spinal ventrolateral pathways.
same spinal regions also to project to more neurons in nuclei usually associated Noxious stimulation evokes prominent signs
caudal brain stem nuclei95,104,105. with affective or motivational reactions. of localized metabolic increases in several
Information projected rostrally by the However, one projection of spinal distinct cerebral cortical areas including the
ventrolateral tracts is complex, and details lamina I to a thalamic ventral medial anterior insula, the anterior cingulate cortex
remain the subject of debate4. Ventrolateral region proposed by Craig and colleagues (ACC) and somatosensory II regions121,122.
projection neurons in spinal lamina I in 1994 has been reported to feature cells Less consistent, more context-influenced or
include selectively nociceptive categories showing selective excitation by localized smaller activations occur in other cortical
conveying activity from a single or several noxious or thermal stimuli. The thalamic regions4,123–126. Mental state, attention and
types of primary afferent nociceptor. Other zone of this projection was reported to be disposition modify the pattern and details
lamina I ventrolateral projection neurons marked by calbindin labelling113; however, of imaged cortical responses to noxious
receive excitation from a combination these findings have been challenged by input121,124,127. There seems to be agreement
of nociceptors and thermoreceptors4. Jones’s laboratory, which questions the that activity in the ACC detected by PET
Ventrolateral projection neurons from distinctiveness of the region defined by and fMRI, evoked by pain-causing stimuli, is
deeper dorsal horn laminae have more calbindin immunocytochemistry114. related to emotion and motivation124,127,128.

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | JANUARY 2007 | 77


© 2007 Nature Publishing Group
PERSPECTIVES

Descending control. Modulation of spinal dissociation from other body sensations in not apply to the chronic situation; however,
mechanisms by activity descending from disorders of the nervous system. However, long-term pain-related activity might be
rostral brain centres was documented in the discomfort or suffering usually related to associated with other profound alterations
Sherrington’s classical neurophysiological the recognition of a painful event are strong in reactions, including sensory experience
studies20. Intrinsic neural mechanisms feelings of the type categorized as emotions. and pathological functions in other organ
controlling pain were given sharp focus by So what is pain? Its ordinary expression has systems. These situations are beyond the
Reynold’s 1969 article describing rodent sur- attributes and an underlying neural organiza- scope of the present perspective.
gery during analgesia produced by electrical tion of both a specific sensation (detecting,
stimulation in the midbrain periaqueductal signalling and recognizing noxious stimuli) Conclusions
grey129. Activation of a number of other and those of an emotion. Should pain be In summary, the past two centuries have
brainstem nuclei produces potent suppres- considered both a specific sense and an emo- seen major alterations in thinking regard-
sion of pain-related spinal activity. This tion? Given these two sides, it is no wonder ing pain, which have paralleled a great
prompted numerous studies of ‘stimulation- that there has been debate and uncertainty expansion in understanding of nervous
produced anaesthesia’ and descending ‘anal- about the nature of pain over the ages. mechanisms. During the last 40 years,
gesia’ pathways86,93. Descending inhibition Modern neuron tracing and human brain incontrovertible evidence has accumulated
of spinal pain-related activity is produced by imaging observations showing connections to show that specialized peripheral sense
neurons utilizing different chemical media- and activity consistent with a discriminative organs act as detectors of tissue-damaging
tors, including serotonin, catecholamines, sense and an affective reaction certainly can events, and that activity in these nocicep-
and endogenous opioids. be argued to reflect such duality. tors normally initiate pain. Their signals
Investigators who concentrate on pain In considering labelled line (that is, have been shown to be partially segregated
have tended to categorize descending systems dedicated sense organs and neural pathways) in transmission to multiple rostral brain
and their effects as specifically anti-nocicep- and convergent-intensity (that is, quantita- centres including the thalamus and the
tive or analgesic arrangements. However, tively controlled, nonspecific neural arrange- cerebral cortex. However, it has become
there is remarkable similarity in the nuclei ments) models for the central organization apparent that activity in neuronal pathways
involved and the mediators employed, of nociceptive and pain systems, Craig characterized by the convergence of primary
between those implicated in analgesic (2003) concluded that pain was a homeo- afferent input has a role in the processes
effects and those involved during sleep static emotion4. However, homeostasis associated with nociception and pain.
and stress64,130. It seems reasonable to view implies holding an organism and its systems Considerable importance has been attached
descending control of spinal reflex and affer- to a stable state, a concept that is difficult to to activity-dependent changes in nocicep-
ent mechanisms in a more general context reconcile with some concomitants of noci- tive neuronal systems and to higher centre
than simply modifying nociception and pain. ception and pain. These include marked control of ascending pathways, even though
increases in respiration, heart rate and the latter might not be unique to pain.
Opioid receptors. Opioid derivatives from arterial pressure. Such functional alterations Finally, current evidence from human imag-
the poppy plant have been used by man for could be appropriate reactions when tissue is ing studies emphasises parallel processing of
centuries, although often not for amelioration injured, but they poorly fit the usual concept sensory information in complex reactions,
of pain. However, morphine and other opiates of homeostasis. including pain.
continue to be extremely important agents for With the benefit of the past two
pain relief. Even though it was long suspected Chronic pain. Present-day clinicians call centuries of scientific work and thought,
that opiates acted on particular receptive pain that persists beyond the usual time can one define pain? Considering the
structures in cells and tissues, an opioid associated with injury of a tissue ‘chronic’, evidence, it seems reasonable to propose
receptor in neural tissue was only definitively often regardless of presumed cause. This pain to be both a specific sensation and an
identified in 1973 by Pert and Snyder131. implies a difference from the usual rela- emotion, initiated by activity in particular
A specialized molecular receptor for opioid tionship between tissue injury and rapid peripheral and central neurons. Pain
compounds strongly suggested the existence pain or nociceptive reactions. There are shares features with other sensations, but
of an endogenous ligand. Shortly after the unquestionable changes in neural function- the strong association with disposition is
identification of opiate receptors, enkephalin ing secondary to persistent nociceptive special. The mechanisms of pain include
was shown by Hughes and Kosterlitz132,133 input. Moreover, long-lasting aberrant specialized receptive organs, selective and
and Simantov and Snyder134 to be one of a CNS-generated activity that triggers the convergent pathways, plasticity of respon-
series of endorphins (endogenous morphine). perception of pain also produces plastic siveness and interactive modulation. No
Activation of opiate receptors by morphine alterations in behaviour and nervous single theory (emotion, specific, intensive,
or synergists produces changes in behaviour function. In addition to phenomena such pattern or gate) fits the present evidence
and suppresses nociceptive reactions, among as sensitization of peripheral nociceptors by itself, although a combination of ideas
other actions. Certain anti-nociceptive opioid or central neurons, rearrangements of answers most issues. Reason suggests an
actions involve the suppression of synaptic activity and possibly neuronal connections integration of features is the best choice for
effectiveness by primary afferent fibres at are reported. Some processes leading to a working hypothesis.
their central terminations. these rearrangements might take place at Department of Cell & Molecular Physiology, 5109D
the genetic level. Principal factors in clini- Neuroscience Research Building, University of North
Sensation–emotion and homeostasis. The cal circumstances are the emotional and Carolina at Chapel Hill, Chapel Hill, North Carolina
primary evidence for considering pain motivational concomitants of persisting 27599-7545, USA.
from noxious stimulation as a discrimina- pain symptoms. This is not to suggest that e-mail: erp@med.unc.edu
tive sense, rather than an emotion, is its the mechanisms discussed in this article do doi:10:1938/nrn2042

78 | JANUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro


© 2007 Nature Publishing Group
PERSPECTIVES

1. Dallenbach, K. M. Pain: history and present status. 31. Heinbecker, P., Bishop, G. H. & O’Leary, J. Pain and 62. Ritter, A. M. & Mendell, L. M. Somal membrane
Am. J. Psychol. 3, 331–347 (1939). touch fibers in peripheral nerves. Arch. Neurol. properties of physiologically identified sensory
2. Keele, K. D. Anatomies of Pain (Charles C. Thomas, Psychiatry 29, 771–789 (1933). neurons in the rat: effects of nerve growth factor.
Springfield, Illinois, 1957). 32. Zotterman, Y. Studies in the peripheral nervous J. Neurophysiol. 68, 2033–2041 (1992).
3. Gruner, O. C. A Treatise on the Canon of Medicine of mechanism of pain. Acta Med. Scand. 80, 1–64 63. Hökfelt, T., Kellerth, J. O., Nilsson, G. & Pernow, B.
Avicenna, Incorporating a Translation of the First Book (1933). Substance P: localization in the central nervous system
(Luzac & Co., London, 1930). 33. Clark, D., Hughes, J. & Gasser, H. S. Afferent and in some primary sensory neurons. Science 190,
4. Craig, A. D. Pain mechanisms: labeled lines versus function in the group of nerve fibers of slowest 889–890 (1975).
convergence in central processing. Annu. Rev. conduction velocity. Am. J. Physiol. 114, 69–76 64. Perl, E. R. in The Nervous System (ed. Darian-Smith, I.)
Neurosci. 26, 1–30 (2003). (1935). 915–975; Series, Handbook of Physiology (eds
5. Bell, C. Idea of a New Anatomy of the Brain; 34. Landau, W. & Bishop, G. H. Pain from dermal, Brookhart, J. M. & Mountcastle, V. B.) (American
Submitted for the Observations of His Friends periosteal, and fascial endings and from inflammation. Physiological Society, Bethesda, Maryland, 1984).
(Strahan and Preston, London, 1811). AMA. Arch. Neurol. Psychiatry 69, 490–504 (1953). 65. Lawson, S. N. in Peripheral Neuropathy (eds Dyck, P. J.
6. Cranefield, P. F. The Way In and the Way Out: Francois 35. Collins, W. F. Jr, Nulsen, F. E. & Randt, C. T. Relation of & Thomas, P. K.) 163–202 (Saunders, Philadelphia,
Magendie, Charles Bell and the Roots of the Spinal peripheral nerve fiber size and sensation in man. Arch. 2005).
Nerves (Futura Publishing Company, Mount Kisco, Neurol. 3, 381–385 (1960). 66. Light, A. R. & Perl, E. R. Spinal termination of
New York, 1974). 36. Lewis, T. & Pochin, E. E. The double pain response of functionally identified primary afferent neurons with
7. Müller, J. Handbuch der Physiologie des Menschen the human skin to a single stimulus. Clin. Sci. 3, slowly conducting myelinated fibers. J. Comp. Neurol.
(J. Holscher, Koblenz, 1840) (in German). 67–76 (1937). 186, 133–150 (1979).
8. Dubois-Reymond, E. Untersuchungen über thierische 37. Adrian, E. D. & Zotterman, Y. The impulses produced 67. Sugiura, Y., Lee, C. L. & Perl, E. R. Central projections
Elektricität (Georg Reimer, Berlin, 1848–1849) (in by sensory nerve endings. Part 2. The response of a of identified, unmyelinated (C) afferent fibers
German). single end-organ. J. Physiol. 61, 11–171 (1926). innervating mammalian skin. Science 234, 358–361
9. Schiff, J. M. Lehrbuch der Physiologie des Menschen. 38. Nafe, J. P. A quantitative theory of feeling. J. Gen. (1986).
Muskel und Nervenphysiologie (Schauenberg, Lahr, Psychol. 2, 199–211 (1929). 68. Echlin, F. & Propper, N. ‘Sensitization’ by injury of the
1858) (in German). 39. Zotterman, Y. Specific action potentials in the lingual cutaneous nerve endings in the frog. J. Physiol. 88,
10. Brown-Séquard, C. E. Course of Lectures on the nerve of cat. Skand. Arch. Physiol. 75, 105–120 388–400 (1936).
Physiology and Pathology of the Central Nervous (1936). 69. Witt, I. & Griffin, J. P. Afferent cutaneous C-fibre
System (Collins, Philadelphia, 1860). 40. Zotterman, Y. Touch, pain and tickling: and reactivity to repeated thermal stimuli. Nature 194,
11. Brown-Séquard, C. E. Lectures on the physiology and electrophysiological investigation on cutaneous 776–777 (1962).
pathology of the nervous system; and on the sensory nerves. J. Physiol. 95, 1–28 (1939). 70. Belmonte, C. & Cervero, F. Neurobiology of
treatment of organic nervous affections. Lancet 2, 41. Sinclair, D. C., Weddell, G. & Zander, E. The Nociceptors (Oxford Univ. Press, Oxford, 1996).
593–596 (1868). relationship of cutaneous sensibility to neurohistology 71. Lewis, T. Pain (Macmillan, New York, 1942).
12. Gowers, W. R. A case of unilateral gunshot injury to in the human pinna. J. Anat. 86, 402–411 (1952). 72. Hardy, J. D., Woolf, H. G. & Goodell, H. Pain
the spinal cord. Trans. Clin. Lond. 11, 24–32 (1878). 42. Weddell, G. Somesthesis and the chemical senses. Sensations and Reactions (Williams and Wilkins,
13. Erb, W. Handbuch der Krankheiten des Ann. Rev. Psychol. 6, 119–136 (1955). Baltimore, Maryland, 1952).
Nervensystems II (F. C. W. Vogel, Leipzig, 1874) 43. Sinclair, D. C. Cutaneous sensation and the doctrine of 73. Schaible, H. G. & Grubb, B. D. Afferent and spinal
(in German). specific energy. Brain 78, 584–614 (1955). mechanisms of joint pain. Pain 55, 5–54 (1993).
14. Blix, M. Experimentelle beiträge zur lösung der frage 44. Lele, P. P. & Weddell, G. The relationship between 74. Schmidt, R. F. & Schaible, H.-G. in Cellular
über die specifische energie der hautnerven. Z. Biol. neurohistology and corneal sensibility. Brain 79, Mechanisms of Sensory Processing (ed. Urban, L.)
20, 141–156 (1884) (in German). 119–154 (1956). 289–296 (Springer, Berlin, 1994).
15. Goldscheider, A. Die specifische energie der 45. Maruhashi, J., Mizuguchi, K. & Tasaki, I. Action 75. Schaible, H.-G. & Schmidt, R. F. in Neurobiology of
gefühlsnerven der haut. Monatshft. Prak. Derm. 3, currents in single afferent nerve fibres elicited by Nociceptors (eds Belmonte, C. & Cervero, F.)
283 (1884) (in German). stimulation of the skin of the toad and the cat. 202–219 (Oxford Univ. Press, Oxford, 1996).
16. von Frey, M. Beiträge zur physiologie des J. Physiol. 117, 129–151 (1952). 76. Dubner, R., Price, D. D., Beitel, R. E. & Hu, J. W. in
schmerzsinns. Königl. Sächs. Ges. Wiss., Math. Phys. 46. Paintal, A. S. Functional analysis of group III afferent Pain in the Trigeminal Region (eds Anderson, D. J. &
Classe 46, 185–196 (1894) (in German). fibres of mammalian muscles. J. Physiol. 152, Matthews, B.) 57–66 (Elsevier/North-Holland
17. von Frey, M. Beiträge zur sinnesphysiologie der haut. 250–270 (1960). Biomedical Press, Amsterdam, 1977).
Dritte mittheilung. Königl. Sächs. Ges. Wiss., Math. 47. Bessou, P. & Laporte, Y. Étude des récepteurs 77. Torebjörk, H. E. Afferent C units responding to
Phys. Classe 47, 166–184 (1895) (in German). musculaires innervés par les fibres afférentes du mechanical, thermal and chemical stimuli in human
18. von Frey, M. Untersuchungen über der groupe III (fibres myelinisées fines) chez le chat. Arch. non-glabrous skin. Acta Physiol. Scand. 92, 374–390
sinnesfunctionen der menschlichen haut. Erste Ital. Biol. 99, 293–321 (1961) (in French). (1974).
ablandlung: druckempfindung und schmerz. Königl. 48. Hunt, C. C. & McIntyre, A. K. Properties of cutaneous 78. Hagbarth, K. E., Hongell, A., Hallin, R. G. &
Sächs. Ges. Wiss., Math. Phys. Classe 48, 175–266 touch receptors in cat. J. Physiol. 153, 88–98 (1960). Torebjörk, H. E. Afferent impulses in median nerve
(1896) (in German). 49. Hunt, C. C. & McIntyre, A. K. An analysis of fibre fascicles evoked by tactile stimuli of the human hand.
19. von Frey, M. Beiträge zur sinnesphysiologie der haut. diameter and receptor characteristics of myelinated Brain Res. 24, 423–442 (1970).
Vierte mittheilung. Königl. Sächs. Ges. Wiss., Math. cutaneous afferent fibers in cat. J. Physiol. 153, 79. Torebjörk, H. E. & Ochoa, J. Specific sensations
Phys. Classe 49, 462–468 (1897) (in German). 99–112 (1960). evoked by activity in single identified sensory units in
20. Sherrington, C. S. The Integrative Action of the 50. Iggo, A. Cutaneous heat and cold receptors with slowly man. Acta Physiol. Scand. 110, 445–447 (1980).
Nervous System (Cambridge Univ. Press, Cambridge, conducting (C) afferent fibres. Quart. J. Exp. Physiol. 80. Konietzny, F., Perl, E. R., Trevino, D., Light, A. &
UK, 1906). 44, 362–370 (1959). Hensel, H. Sensory experiences in man evoked by
21. Waldeyer, W. Über einige neurere forschungen in 51. Iggo, A. Cutaneous mechanoreceptors with afferent C intraneural electrical stimulation of intact cutaneous
gebiete der anatomie des centralnervensystems. fibres. J. Physiol. 152, 337–353 (1960). afferent fibers. Exp. Brain Res. 42, 219–222 (1981).
Deutsche Med. Wochenschr. 44, 1–64 (1891) (in 52. Iriuchijima, J. & Zotterman, Y. The specificity of 81. Ochoa, J. & Torebjörk, E. Sensations evoked by
German). afferent cutaneous C fibres in mammals. Acta. Physiol. intraneural microstimulation of single
22. Edinger, L. Einigles von verlauf der gefühlsbahnen in Scand. 49, 267–278 (1960). mechanoreceptor units innervating the human hand.
centralen nervensysteme. Deutsche Med. Woch. 16, 53. Melzack, R. & Wall, P. D. On the nature of cutaneous J. Physiol. 342, 633–654 (1983).
421–426 (1890) (in German). sensory mechanisms. Brain 85, 331–356 (1962). 82. Wall, P. D. & McMahon, S. B. Microneurography and
23. Edinger, L. Zwölf Vorlesungen über den Bau der 54. Melzack, R. & Wall, P. D. Pain mechanisms: a new its relation to perceived sensation. A critical review.
Nervösen Centralorgane. Für Ärzte und Studirende theory. Science 150, 971–979 (1965). Pain 21, 209–229 (1985).
150–153 (F. C. W. Vogel, Leipzig, 1892). 55. Eccles, R. M. & Lundberg, A. Supraspinal control of 83. Torebjörk, H. E., Vallbo, Å. B. & Ochoa, J. L.
24. Bechterew, V. M. Les Voies de Conduction du Cerveau interneurones mediating spinal reflexes. J. Physiol. Intraneural microstimulation in man. Its relation to
et de la Moelle (A. Maloine, Paris, 1900) (in French). 147, 565–584 (1959). specificity of tactile sensations. Brain 110,
25. Spiller, W. G. & Martin, E. The treatment of 56. Kuno, M. & Perl, E. R. Alteration of spinal reflexes by 1509–1529 (1987).
persistent pain of organic origin in the lower part of interaction with suprasegmental and dorsal root 84. Christensen, B. N. & Perl, E. R. Spinal neurons
the body by division of the anterolateral column of activity. J. Physiol. 151, 103–122 (1960). specifically excited by noxious or thermal stimuli:
the spinal cord. J. Am. Med. Assoc. 58, 1489–1490 57. Burgess, P. R. & Perl, E. R. Myelinated afferent fibres marginal zone of the dorsal horn. J. Neurophysiol. 33,
(1912). responding specifically to noxious stimulation of the 293–307 (1970).
26. Dejerine, J. & Roussy, G. Le syndrome thalamique. skin. J. Physiol. 190, 541–562 (1967). 85. Kumazawa, T. & Perl, E. R. Excitation of marginal and
Rev. Neurol. (Paris) 14, 521–532 (1906) (in French). 58. Perl, E. R. Myelinated afferent fibres innervating the substantia gelatinosa neurons in the primate spinal
27. Head, H. & Holmes, G. Sensory disturbances from primate skin and their response to noxious stimuli. cord: indication of their place in dorsal horn functional
cerebral lesions. Brain 34, 102–254 (1911). J. Physiol. 197, 593–615 (1968). organization. J. Comp. Neurol. 177, 417–434
28. Head, H., Rivers, W. H. R. & Sherren, J. The afferent 59. Bessou, P. & Perl, E. R. Response of cutaneous (1978).
nervous system from a new aspect. Brain 28, 99–115 sensory units with unmyelinated fibers to noxious 86. Light, A. R. The Initial Processing of Pain and its
(1905). stimuli. J. Neurophysiol. 32, 1025–1043 (1969). Descending Control: Spinal and Trigeminal Systems
29. Ranson, S. W. Unmyelinated nerve-fibres as 60. Bessou, P., Burgess, P. R., Perl, E. R. & Taylor, C. B. (ed. Reichmann, H.) (S. Karger AG, Basel, 1992).
conductors of protopathic sensation. Brain 38, Dynamic properties of mechanoreceptors with 87. Foerster, O. & Gagel, O. Die vorderseitenstrangdurch-
381–389 (1915). unmyelinated (C) fibers. J. Neurophysiol. 34, schneidung beim menschen. Eine klinisch-patho-
30. Erlanger, J., Gasser, H. S. & Bishop, G. H. The 116–131 (1971). physiologische-anatomische studie. Z. ges. Neurol.
compound nature of the action current of nerve as 61. Caterina, M. J. et al. The capsaicin receptor: a heat- Psychiat. 138, 1–92 (1932) (in German).
disclosed by the cathode ray oscillograph. Am. J. activated ion channel in the pain pathway. Nature 88. Kuru, M. The Sensory Paths in the Spinal Cord and
Physiol. 70, 624–666 (1924). 389, 816–824 (1997). Brain Stem of Man (Sogensya, Tokyo, 1949).

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | JANUARY 2007 | 79


© 2007 Nature Publishing Group
PERSPECTIVES

89. Willis, W. D., Kenshalo, D. R. Jr & Leonard, R. B. The 108. Al-Chaer, E. D., Lawand, N. B., Westlund, K. N. & 126. Brooks, J. C., Nurmikko, T. J., Bimson, W. E.,
cells of origin of the primate spinothalamic tract. Willis, W. D. Visceral nociceptive input into the ventral Singh, K. D. & Roberts, N. fMRI of thermal pain:
J. Comp. Neurol. 188, 543–573 (1979). posterolateral nucleus of the thalamus: a new function effects of stimulus laterality and attention.
90. Mendell, L. M. Physiological properties of for the dorsal column pathway. J. Neurophysiol. 76, Neuroimage 15, 293–301 (2002).
unmyelinated fiber projection to the spinal cord. Exp. 2661–2674 (1996). 127. Davis, K. D., Taylor, S. J., Crawley, A. P., Wood, M. L. &
Neurol. 16, 316–332 (1966). 109. Hitchcock, E. Stereotactic cervical myelotomy. Mikulis, D. J. Functional MRI of pain- and attention-
91. Mayer, D. J., Price, D. D. & Becker, D. P. J. Neurol. Neurosurg. Psychiatry 33, 224–230 (1970). related activations in the human cingulate cortex.
Neurophysiological characterization of the 110. Burstein, R., Cliffer, K. D. & Giesler, G. J. Jr. Direct J. Neurophysiol. 77, 3370–3380 (1997).
anterolateral spinal cord neurons contributing to pain somatosensory projections from the spinal cord to the 128. Derbyshire, S. W. G., Vogt, B. A. & Jones, A. K. P. Pain
perception in man. Pain 1, 51–58 (1975). hypothalamus and telencephalon. J. Neurosci. 7, and stroop interference tasks activate separate
92. Price, D. D. & Mayer, D. J. Neurophysiological 4159–4164 (1987). processing modules in anterior cingulate cortex. Exp.
characterization of the anterolateral quadrant neurons 111. Bernard, J. F. & Besson, J. M. The spino(trigemino) Brain Res. 118, 52–60 (1998).
subserving pain in M. mulatta. Pain 1, 59–72 (1975). pontoamygdaloid pathway: electrophysiological 129. Reynolds, D. V. Surgery in the rat during electrical
93. Willis, W. D. The Pain System (Karger, Basel, 1985). evidence for an involvement in pain processes. J. analgesia induced by focal brain stimulation. Science
94. Perl, E. R. in Somatosensory Mechanisms (eds Neurophysiol. 63, 473–490 (1990). 164, 444–445 (1969).
Euler, C. N., Franzin, O., Lindblom, U. & Ottoson, J. D.) 112. Cliffer, K. D., Burstein, R. & Giesler, G. J. Jr. 130. Mason, P. Deconstructing endogenous pain
1–21 (Plenum, New York, 1984). Distributions of spinothalamic, spinohypothalamic, modulations. J. Neurophysiol. 94, 1659–1663
95. Willis, W. D. & Westlund, K. N. Neuroanatomy of the and spinotelencephalic fibers revealed by anterograde (2005).
pain system and of the pathways that modulate pain. transport of PHA-L in rats. J. Neurosci. 11, 852–868 131. Pert, C. B. & Snyder, S. H. Opiate receptor:
J. Clin. Neurophysiol. 14, 2–31 (1997). (1991). demonstration in nervous tissue. Science 179,
96. Dostrovsky, J. O. in Nervous System Plasticity and 113. Craig, A. D., Bushnell, M. C., Zhang, E. T. & 1011–1014 (1973).
Chronic Pain (eds Sandkuhler, J., Bromm, B. & Blomqvist, A. A thalamic nucleus specific for pain and 132. Hughes, J. Isolation of an endogenous compound from
Gebhart, G. F.) 245–258 (Elsevier, Amsterdam, 2000). temperature sensation. Nature 372, 770–773 (1994). the brain with pharmacological properties similar to
97. Woolf, C. J. Evidence for a central component of post- 114. Graziano, A. & Jones, E. G. Widespread thalamic morphine. Brain Res. 88, 295–308 (1975).
injury pain hypersensitivity. Nature 306, 686–688 terminations of fibers arising in the superficial medullary 133. Kosterlitz, H. W. & Hughes, J. Some thoughts on the
(1983). dorsal horn of monkeys and their relation to calbindin significance of enkephalin, the endogenous ligand. Life
98. Ji, R. R., Kohno, T., Moore, K. A. & Woolf, C. J. Central immunoreactivity. J. Neurosci. 24, 248–256 (2004). Sci. 17, 91–96 (1975).
sensitization and LTP: do pain and memory share 115. Head, H. Studies in Neurology (Oxford Univ. Press, 134. Simantov, R. & Snyder, S. H. Morphine-like peptides in
similar mechanisms? Trends Neurosci. 26, 696–705 London, 1920). mammalian brain: isolation, structure elucidation, and
(2003). 116. Penfield, W. & Boldrey, E. Somatic motor and sensory interactions with the opiate receptor. Proc. Natl Acad.
99. Lever, I. J. et al. Brain-derived neurotrophic factor is representation in the cerebral cortex of man as studied Sci. 73, 2515–2519 (1976).
released in the dorsal horn by distinctive patterns of by electrical stimulation. Brain 60, 389–443 (1937). 135. Gasser, H. S. & Erlanger, J. The role played by the
afferent fiber stimulation. J. Neurosci. 21, 117. Dejerine, J. & Mouzon, J. Un nouveau type de sizes of the constituent fibers of a nerve trunk in
4469–4477 (2001). syndrome sensitif cortical observé dans un cas de determining the form of its action potential wave. Am.
100. Mantyh, P. W. et al. Receptor endocytosis and monoplégie corticale dissociés. Rev. Neurol. 18, J. Physiol. 80, 522–547 (1927).
dendrite reshaping in spinal neurons after 1265–1273 (1915) (in French). 136. Erlanger, J. & Gasser, H. S. The action potential in
somatosensory stimulation. Science 268, 118. Kleist, K. in Handbuch der Artzlichen Erfahrungen im fibers of slow conduction in spinal roots and somatic
1629–1632 (1995). Weltkriege 1914–1918 Geistesund nerves. Am. J. Physiol. 92, 43–82 (1930).
101. Trevino, D. L., Maunz, R. A., Bryan, R. N. & Willis, W. D. Nervenkrankheiten (ed. von Schjerning, O.) 137. Erlanger, J. & Gasser, H. S. Electrical Signs of Nervous
Location of cells of origin of the spinothalamic tract in 343–1393 (Barth, Leipzig, 1934). Activity (Univ. Pennsylvania Press, Philadelphia, 1937).
the lumbar enlargement of cat. Exp. Neurol. 34, 119. Russell, W. R. Transient disturbances following 138. Gasser H. S. The classification of nerve fibres. Ohio J.
64–77 (1972). gunshot wounds of the head. Brain 68, 6–97 (1945). Sci. 41, 145–159 (1941).
102. Trevino, D. L. & Carstens, E. Confirmation of the 120. Marshall, J. Sensory disturbances in cortical wounds 139. Gasser H. S. & Grundfest H. Axon diameters in
location of spinothalamic neurons in the cat and with special reference to pain. J. Neurol. Neurosurg. relation to the spike dimension and the conduction
monkey by the retrograde transport of horseradish Psychiatry 14, 187–204 (1951). velocity in mammalian A fibres. Am. J. Physiol. 127,
peroxidase. Brain Res. 98, 177–182 (1975). 121. Derbyshire, S. W. G. et al. Pain processing during 393–414 (1939).
103. Dilly, P. N., Wall, P. D. & Webster, K. E. Cells of origin three levels of noxious stimulation produces 140. Kumazawa, T. & Perl, E. R. Primate cutaneous sensory
of the spinothalamic tract in the cat and rat. Exp. differential patterns of central activity. Pain 73, units with unmyelinated (C) afferent fibres.
Neurol. 21, 550–562 (1968). 431–445 (1997). J. Neurophysiol. 40, 1325–1338 (1977).
104. Mehler, W. R. in Basic Research in Paraplegia (eds 122. Derbyshire, S. W. G. & Jones, A. K. P. Cerebral
French, J. D. & Porter, R. W.) 26–55 (Charles C. responses to a continual tonic pain stimulus measured Acknowledgements
Thomas, Springfield, Illinois, 1962). using positron emission tomography. Pain 76, I am very grateful to B. Taylor-Blake for her invaluable help
105. Mehler, W. R., Feferman, M. E. & Nauta, W. J. H. 127–135 (1998). with the bibliography and preparation of the manuscript.
Ascending axon degeneration following anterolateral 123. Tommerdahl, M., Delemos, K. A., Vierck, C. J. Jr, Support for the preparation of this article was provided by a
cordotomy. An experimental study in the monkey. Favorov, O. V. & Whitsel, B. L. Anterior parietal cortical grant from the National Institutes of Health.
Brain 83, 718–750 (1960). response to tactile and skin-heating stimuli applied to
106. Hirshberg, R. M., Al-Chaer, E. D., Lawand, N. B., the same skin site. J. Neurophysiol. 75, 2662–2670 Competing interests statement
Westlund, K. N. & Willis, W. D. Is there a pathway in (1996). The author declares no competing financial interests.
the posterior funiculus that signals visceral pain? Pain 124. Rainville, P., Duncan, G. H., Price, D. D., Carrier, B. &
67, 291–305 (1996). Bushnell, M. C. Pain affect encoded in human anterior
107. Al-Chaer, E. D., Lawand, N. B., Westlund, K. N. & cingulate but not somatosensory cortex. Science 277, FURTHER INFORMATION
Willis, W. D. Pelvic visceral input into the nucleus 968–971 (1997). Perl’s homepage:
gracilis is largely mediated by the postsynaptic dorsal 125. Craig, A. D., Chen, K., Bandy, D. & Reiman, E. M. http://www.med.unc.edu/physiolo/fac_perl.htm
column pathway. J. Neurophysiol. 76, 2675–2690 Thermosensory activation of insular cortex. Nature Access to this links box is available online.
(1996). Neurosci. 3, 184–190 (2000).

80 | JANUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro


© 2007 Nature Publishing Group

You might also like