You are on page 1of 10

Review Article

Page 1 of 10

Pathophysiology of pain
Sheng-Xi Wu, Ya-Yun Wang, Juan Shi, Yu-Lin Dong, Guo-Hong Cai

Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032,
China
Correspondence to: Sheng-Xi Wu, MD, PhD, Professor; Ya-Yun Wang, MD, PhD, Associate Professor; Juan Shi, PhD, Associate Professor; Yu-Lin
Dong, MD, PhD, Associate Professor; Guo-Hong Cai, Postgraduate Student. Department of Anatomy, Histology and Embryology, K. K. Leung
Brain Research Centre, The Fourth Military Medical University, No. 169 West Changle Road, Xi’an 710032, China. Email: xym@amegroups.com.

Abstract: Pain can be defined as an unpleasant sensory and emotional experience of actual or potential
tissue damage or an experience expressed in such terms. It is a major symptom of many different diseases and
a subjective result of nociception, and can be classified in various ways such as by physiological, etiology and
temporal. Pathophysiological mechanisms including pain pathways, peripheral and central mechanisms that
are involved in pain will be described in this article.

Keywords: Pathophysiology; pain; mechanism

Received: 08 June 2016; Accepted: 23 June 2016; Published: 29 June 2016.


doi: 10.21037/xym.2016.06.05
View this article at: http://dx.doi.org/10.21037/xym.2016.06.05

Introduction contains acute and chronic pain (2).


Acute pain often lasts less than 3 months (3), and is often
Pain can be defined as an unpleasant sensory and emotional
associated with an identifiable injury or trauma. Thus, it
experience of actual or potential tissue damage or an
responds to relatively simple analgesic therapy, and it will
experience expressed in such terms. Both excitatory
be resolved as the area of injury heals or is treated. Acute
and inhibitory physiologic and pathologic mechanisms
physiological nociceptive pain is elicited when a noxious
are involved in its generation and maintenance. Recent
stimulus is applied to normal tissue. Because withdrawal
research has uncovered important pathophysiological
mechanisms that are underlying different clinically relevant reflexes are usually elicited, this pain would protect tissue
pain disorders. This chapter will briefly describe types of from being further damaged. When the tissue is inflamed
pain, pain pathways, peripheral and central mechanisms or injured, pathophysiologic nociceptive pain will occur,
that are involved in pain. A better understanding of which is one of the most frequent reasons why patients seek
pain, particularly the translation of pathophysiological medical care. This kind of pain may appear as spontaneous
mechanisms into sensory signs, will lead to a more effective pain, hyperalgesia, and/or allodynia (4).
and specific mechanism-based treatment approach. Chronic pain lasts longer than 3 months and exceeds
the typical recovery time for the initial injury. It may or
may not be explained by low levels of identified underlying
Types of pain pathology (1). It is often accompanied by neuroendocrine
Pain is a major symptom of many different diseases and a dysregulation, fatigue, increased irritability, depression,
subjective result of nociception. Nociception is the encoding social withdrawal, emotional stress, loss of libido, disturbed
and processing of noxious stimuli in the nervous system that sleep patterns and weight loss (5). Although injury may
can be measured with electrophysiological techniques (1). elicit chronic pain, factors remote from its cause may
Pain may be classified in various ways: physiologically, it perpetuate it. Environmental and affective factors can also
contains nociceptive and neuropathic pain; by etiology, it exacerbate and perpetuate chronic pain. Interestingly, the
contains malignant and nonmalignant pain; temporally, it causal relationship between nociception and pain may not

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Page 2 of 10 Xiangya Medicine, 2016

be clear in many chronic pain states and certain pain states physical insult, and the severity of pain may be much greater
may not reflect tissue damage. than the extent of damage might suggest. Although the
mechanism underlying neuropathic pain is not completely
understood, it is considered to be complex, multifactoral,
Chronic pain
and could evolve over time. Normally, peripheral and central
Chronic pain may also be categorized by its character. It can sensitization phenomena would dissipate as tissue heals and
be nociceptive, neuropathic, or both. It may be caused by inflammation subsides. However, neuropathic pain may be
cancer, inflammation, or a variety of non-life-threatening elicited when changes in primary afferent function persist
conditions such as arthritis, fibromyalgia, and neuropathy (2). after disease or injury of the nervous system (9).
Chronic pain may result from a chronic disease and then
actually result from persistent nociceptive processes.
Inflammation pain

Chronic inflammation is a feature of a large number of painful


Cancer pain
chronic degenerative diseases, such as arthritis, low back
Most chronic cancer-related pains are caused directly by the pain, and inflammatory bowel disease (11). Unfortunately, it
tumor. Bone metastases and compression of neural structures may be the single greatest cause of pain, which is the result of
are the two most common causes (6). Bone metastases could activation and sensitization of primary afferent nerve fibers.
potentially cause pain by many mechanisms, including Inflammation pain is experienced predominantly when the
endosteal or periosteal nociceptor activation or tumor growth inflamed site is mechanically stimulated by being moved
into adjacent soft tissue and nerves (7). In fact, there is no or touched. The stimulation-induced pain is thought to
homogenous entity of “cancer pain”; pain in cancer, which be referred to as inflammatory hyperalgesia. Inflammatory
is often tied up with concomitant psychosocial upheaval and hyperalgesia has been shown to be produced by inflammatory
existential anxiety, can be inflected by a multitude of emotional, mediators released from circulating leucocytes and platelets,
psychological and spiritual factors (8). To many cancer vascular endothelial cells, immune cells resident in tissue and
sufferers, their pain has a “sinister meaning” over and above its sensory and sympathetic nerve fibers. Since the first mediator
inherent unpleasantness as a sensory experience (8). Chronic bradykinin was reported in 1953 (12), researchers have
cancer pain rarely involves medicolegal or disability issues and identified a number of inflammatory mediators which can
is distinct from acute physiological pain. It has a significant produce hyperalgesia, including prostaglandins, leukotriene,
effect on every aspect of a person’s life, including daily physical serotonin, adenosine, histamine, interleukin-1, interleukin-8
activities, mood, social relationships, sleep patterns, cognition and nerve growth factor (NGF).
and existential beliefs (whether the person’s life has meaning,
purpose or value in relation to mortality) (2). The patients with
Pathways for pain
cancer may become withdrawn and unable to focus if the pain
is not managed well. Pain is a sensation that evokes an emotional response and
involves a complex interaction between the periphery, the
spinal cord, the brainstem and higher cortical centers.
Neuropathic pain

Neuropathic pain is thought to arise from abnormal physiology


Nociceptors
of the peripheral or central nervous systems and may be
unrelated to ongoing tissue damage or inflammation (9). Nociceptive afferents do not have specialized receptors;
There are numerous causes of nervous system injury, they use free nerve endings and most are polymodal (13).
including exposure to toxins, infection, viruses, metabolic They respond to more than one kind of stimulus, such as
disease, nutritional deficiencies, ischemia, trauma (surgical chemical, thermal or mechanical stimuli. Free nerve endings
and nonsurgical), and stroke (10). Some patients with are found in all parts of the body except the interior of the
neuropathic pain may have severe pain without obvious bones and the brain itself. Aδ fibers mediate the “fast” pain,
clinical signs of nerve injury, whereas others may have whereas C-fibers signal the “slow” pain. Not all Aδ and
significant nerve injury without pain. In addition, C fibers are nociceptors. Some respond to low threshold
neuropathic pain may be precipitated by a relatively minor stimuli such as sensual touching or brushing the skin. Many

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Xiangya Medicine, 2016 Page 3 of 10

C fibers are thermoreceptors, and respond to warm or cold, reticular formation. The slow pain axons innervate the
providing homeostatic responses via emotional tagging of non-specific intrathalaminar nuclei of the thalamus, and
sensations. Additionally, there is a population of “silent” the autonomic centers of the reticular formation in the
nociceptors that reside in most tissues and are normally brainstem. Slow pain may remind the brain that pain has
insensitive to mechanical and thermal stimuli. They become occurred, that protective attention to the injury site is
active under pathological conditions such as inflammation required and that normal activity may need to be restricted
and nerve injury. while healing occurs (20).
There are several differences between cutaneous and The projections to the reticular formation underlie the
muscle or visceral nociceptors (14-16). The first is that arousal effects of the painful stimuli via activation of the
cutaneous sensation is well localized and the pain is usually ascending reticular activating system that projects to all
constant. Visceral and muscle pain is poorly localized, due areas of the brain. Activation of the reticular formation
to the lower innervation densities of these tissues, and is stimulates noradrenergic neurons in the locus coeruleus,
often periodic. Secondly, visceral afferents are insensitive and thus decreases the pain transmission by activating the
to direct trauma but very sensitive to distension. Lastly, descending pain modulating system (21).
as most visceral organs have very few low threshold Thalamocortical axons transmit the information from
myelinated fibers and comprise mostly of Aδ and C fibers, the thalamus to the cortex. There is no one specific cortical
their stimulus response properties differ from cutaneous region that can be designated as “pain cortex”. Rather,
and muscle afferents, which have specialized receptors functional brain-imaging studies have revealed several
to detect innocuous stimuli. Visceral afferents encode a regions that are active when a pain stimulus is sensed and
stimulus response in an intensity dependent manner, the these are associated with different functional components
more painful the stimulus the greater the number of action of pain (22,23). “Where and how much it hurts” involves
potentials and frequency of discharge. the somatosensory cortex whereas “I do not like it or stop
it” are associated with the limbic regions, such as cingulate
cortex or insula. Parts of the prefrontal motor cortex are
Cutaneous pain pathways
also involved in cognitive evaluative process.
Cutaneous nociceptor afferents terminate mainly in
laminae I, II and V of the spinal cord dorsal horn and
Visceral and muscular pain pathways
synapse on second order neurons that carry the signal to
either the brainstem or the thalamus. It is only when the Considering that muscle and visceral pain evoke distinct
nociceptive signal reaches the brainstem that it is translated sensations, it would be logical to expect to find cells in
into a conscious sensory perception. Three ascending the spinal cord that respond only to muscle or visceral
pathways are concerned with pain transmission: the stimulation. Interestingly, no such cells exist in the spinal
spinothalamic tract (STT), the spinoreticular tract (SRT) cord. All cells that have either a visceral or muscle receptive
and the spinoparabrachial tract (SPBT). Each appears to field also have a separate cutaneous receptive field. This
be concerned with a particular aspect of pain processing. means that convergence occurs within the spinal cord.
Simplistically, the sensory discriminative aspect is signaled The central terminals of visceral and muscle nociceptors
by the STT, and the homeostatic and affective qualities of terminate in laminae I and V but also laminae II, unlike
pain are signaled by the SRT and SPBT (17-19). Therefore, skin nociceptor afferents. In lamina I, these fibers converse
fast and slow pain travel by different pathways to different onto projection neurons of the STT and SPBT, which then
areas of the brain. The fast pathway connects directly to the project to the brainstem and thalamus and from there to
thalamus, which then relays the information to the primary the somatosensory cortex (24,25). Visceral afferents also
sensorimotor cortices for analysis and response. Its function terminate on SPT neurons and onto cells that project to
is to act as a warning system by signaling the exact location the dorsal column nuclei and recent research suggests that
and severity of the injury and the duration of the pain. this latter pathway is exclusively involved in visceral pain,
Fast pain predominantly arises from STT neurons in the whereas the STT and SRT visceral pathways are more
laminae IV–V of the spinal cord. Slow pain is mediated by concerned with autonomic reflex functions than visceral
C-fibers and signals the emotional aspects of pain. It reaches pain (26). Lesions of the dorsal columns effectively relieve
the thalamus indirectly via connections with the brainstem chronic visceral pain and have led to new clinical treatments

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Page 4 of 10 Xiangya Medicine, 2016

for managing visceral cancer pain. the majority of noradrenergic neurons with projections
to SDH (37). These supraspinal structures together form
the descending control system of the spinal nociceptive
Spinal dorsal horn loco circuits for pain modulation
transmission.
The initial integration of noxious information occurs in Complicated circuits can be formed between those
the superficial dorsal horn (SDH) of the spinal cord, which key elements within SDH, thus facilitating or inhibiting
is very important for understanding pain sensation and nociceptive transmission. These circuits can be delineated
developing effective analgesic strategies (27). according to different criteria. However, they are not hard-
Peripheral nociceptors transmit noxious information wired since they are capable of dynamic change under
to spinal neurons that further project afferents through different pain states. Furthermore, depending on the
ascending tracts, such as the STT, and neurons located in membrane properties (e.g., different receptor types on the
the SDH (including laminae I and II). These secondary and membrane), the same classic neurotransmitter can function
third order axons then synapse with sensory relay nuclei inhibitorily or excitatorily.
in the thalamus and then onto the cerebral cortex. SDH
neurons (including projection neurons and interneurons)
Peripheral mechanisms of pain
receive nociceptive information conveyed by the thinly
myelinated (Aδ) and unmyelinated (C) primary afferent At the peripheral level, a “pain” signal is initiated when
fibers (PAFs) (28,29). Additionally, SDH neurons and PAFs physicochemical stimuli of high intensity or a particular
receive dense descending inputs, including serotonergic quality are detected by a subpopulation of sensory
and noradrenergic components mainly originating from neurons termed nociceptors. Traditionally, nociceptors are
the raphe magnus and locus ceruleus in the brain stem noncapsulated nerve endings of thin myelinated (Aδ-type)
respectively (30). The sole noxious output of SDH and nonmyelinated (C-type) fibers in the tissue, which can
projection neurons to the higher brain structures are likely sense the actual or potential tissue damage and convert
modulated by complicated interneuronal interactions within the energy into a changed membrane potential that is
the SDH. Thus the integration of nociceptive transmission sufficient to generate an action potential, a cognizable
within the SDH is very important for understanding pain “language” of nervous system. Most of the nociceptors
sensation and developing effective analgesic strategies. The respond to mechanical, chemical and/or cold stimuli and
platform for such complicated integrations is the circuits thus are polymodal. During the pathophysiological process,
formed by pools of neurons that are highly plastic under however, the nociceptor per se also undergoes functional or
conditions of inflammatory or neuropathic pain. structural changes in response to the tissue damage. These
changes may amplify the effect of initial stimulus to make
pain either a powerful defense system for individual safety
Descending pain control pathways
(nociceptive pain) or even a diseased condition torturing
The idea of descending control systems dates back to patients (chronic pain). Due to the limited space, we will
the 1970s and 1980s when its original version was that focus on reviewing the main peripheral mechanisms of pain,
structures in the brain stem send impulses that ‘‘descend’’ which have been proved to be effective targets for clinical
and thus inhibit the nociceptive transmission in SDH therapy.
(31,32). A simplified descending inhibitory system was
depicted as the following: the PAG projects to the rostral
Peripheral sensitization
ventromedial medulla (RVM), which includes the serotonin-
rich nucleus raphe magnus (NRM) as well as the nucleus During tissue injury or inflammation, polymodal nociceptors
reticularis gigantocellularis pars alpha and the nucleus undergo decreased excitation thresholds, increased
paragigantocellularis lateralis (33). Serotonergic (34) and response to suprathreshold stimuli, such that even light
some GABAergic or glycinergic (35,36) neurons in RVM or normally innocuous stimuli can activate the receptors.
then project along DLF to terminate in SDH, where Such phenomenon is referred to as peripheral sensitization
they inhibit nociceptive transmission. Besides RVM, or nociceptor sensitization (38). As a consequence, noxious
locus coeruleus and subcoeruleus nucleus are important stimuli applied to the injured tissue area evoke more severe
supraspinal descending inhibitory structures which contain pain (primary hyperalgesia) than in the non-sensitized state

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Xiangya Medicine, 2016 Page 5 of 10

and innocuous stimuli such as mechanical and cold stimuli referred to as ectopic discharge (ectopia) (42). A feature
also cause pain (allodynia). Thus, peripheral sensitization is of ectopic discharge is its mechanosensitivity, e.g., gentle
a neuronal mechanism correlating with behavioral changes, and instantaneous touch of the injured nerves or spinal
i.e., hyperalgesia and allodynia. In addition to triggering the roots generates intense and outlasting discharges, which
increased sensitivity of nociceptor to stimuli, inflammation causes the sensation reminiscent of the stabbing or shock
also causes the recruitment of so-called “silent nociceptors”. like sensation in patients with neuropathic pain. Thus, it
Silent nociceptors are A- or C-fibers that are unexcitable is currently regarded that ectopic discharges of differential
or hard to excite with a high mechanical threshold under afferent fibers likely underlie the clinical manifestations
healthy conditions. During inflammation, however, these called burning pain (C-fiber), dysesthesias (Aδ-fiber) or
fibers are sensitized and become susceptible to applied paresthesias (Aδ-fiber) resulting from nerve injury (10).
stimuli (39). Interestingly, a growing body of evidence suggests that the
Nociceptor sensitization is initiated by the action intact nerve neighboring injured axons may also develop
of numerous inflammatory mediators released upon ectopic activities (43).
tissue injury from surrounding cells such as mast cells, Ectopia, which is characterized by tonic, burstic or
keratinocytes, macrophages and immune cells (4). Important irregular firing pattern, results from secondary changes
among these inflammatory mediators or cytokines are that develop in hours and days following nerve injury (42).
bradykinin, serotonin, histamine, prostaglandin, adenosine, The cellular mechanisms of ectopic impulse generation is
interleukins, tumor necrosis factor alpha as well as not simply a decreased spike threshold but rather complex
neurotrophins like NGF (40). A mixture of these substances alterations in DRG neurons involving gene transcription,
forms the “sensitizing soup” and modulates the sensitivity protein trafficking and ion channel kinetics.
of nociceptors to thermal, mechanical or chemical stimuli. One plausible example for these changes in DRG neurons
Primary afferent neurons express receptors for all these is altered expression and location of Na+ channels. After
substances (however not all receptors are expressed in nerve injury, Nav1.3 and Nav2 increased while Nav1.1, 1.2,
each nociceptor). The binding of specific ligands to these 1.6, 1.7, 1.8, 1.9 decreased in primary afferent neurons (44).
receptors leads to either the activation of nociceptors The distribution of Na+ channels also tends to accumulate
directly or indirectly via second messenger cascades which in neuroma endbulbs and sprouts, rather than in the sensory
in turn influence the functional state or the expression of nerve endings and intra-segmental parts of the node of
ion channels in the cell membrane (38). Through such Ranvier. The kinetics of Na+ channels are also modulated
exogenous processes, the excitability of primary afferent by proinflammatory cytokines and other downstream
neurons can be greatly enhanced with lowered threshold mediators such as protein kinases (PKA and PKC) (44). In
and increased action potential frequency by suprathreshold addition to Na+ channels, N-type Ca2+ channels are also
stimulation. Nerve cells are highly innervating cells at found to decrease in expression in DRG neurons (42).
any time; in addition to passive alteration by micro-milieu All these quantitative and qualitative changes of channel
substances, they are able to release neuropeptides from their proteins contribute to the altered electrophysiological
peripheral terminals. The typical examples are calcitonin- phenotype of injured nociceptive neurons. In support
gene related peptide (CGRP) and substance P (SP). CGRP of this, some clinically effective analgesics such as local
causes vasodilation and SP induces protein extravasation, anesthetic lidocaine and anticonvulsant gabapentin actually
both effects being deteriorating inflammatory processes (41). act as modulators of Na+ and Ca2+ channels, respectively (45).

Ectopic discharge Sympathetically maintained pain

As mentioned above, sensory discharges originate from Spinal nerve comprises four types of nerve fibers, i.e.,
nerve endings in somatic and visceral tissues where the somatic afferent and efferent, and visceral afferent and
receptor potential is transduced into action potential. When efferent (sympathetic or parasympathetic) fibers. Normally,
injury occurs to peripheral nerves, however, other cellular the sympathetic nervous system does not activate somatic
compartments such as the proximal parts of injury and the primary afferents, while in the nerve-injured conditions,
dorsal root ganglia (DRG) cell bodies turn to generate such cross-talk might occur. The responsiveness of afferent
action potential, an electrophysiological phenomenon neurons to sympathetic transmitters (noradrenaline and

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Page 6 of 10 Xiangya Medicine, 2016

adrenaline) is likely to contribute to causalgia, the burning central sensory neurons exist and peripheral noxious
pain in patients with injury in the main nerve of a limb inputs are transmitted. In chronic pain states, modification
(46-48). This is evidenced by the effectiveness of systemic and plasticity of local neural circuits in the spinal dorsal
injection of phentolamine, the α-adrenoceptor blocker, horn (SDH) were induced. As a result, ongoing noxious
in alleviating the pain (49). Such pain depending on the information to the brain is amplified and prolonged, a
activity of sympathetic neurons is termed sympathetically phenomenon which we call central sensitization. Central
maintained pain (SMP). SMP is a symptom characterized sensitization is the abnormal hyperexcitability of nociceptive
by spontaneous pain and/or pain evoked by mechanical or related neurons in the SDH following intense peripheral
thermal stimuli and may be present in the complex regional noxious stimuli, tissue injury or nerve damage (54-56).
pain syndrome (CRPS) type I and II (50). Nociceptive information is transmitted from peripheral sites
The mechanisms for SMP are not well understood to the SDH via unmyelinated (type C) and myelinated (type
despite long exhaustive efforts of researchers over tens of Aδ) fibers. Usually, central sensitization disappears when
years. Several experimental models are, however, suggestive the peripheral input is reduced. However, in some serious
and influential. Following nerve section or ligation, pathological pain, central sensitization can be isolated
electrical stimulation of lesioned or intact sympathetic from the peripheral nociceptive transmission. At this time,
supply excites unmyelinated and myelinated afferent fibers. the central sensitization exists even when the peripheral
This coupling is chemical and may occur via noradrenaline stimulation and inputs disappear. Processes that are known
acting on the α2-adrenoceptors, as the ephaptic coupling to contribute to hyperexcitability of dorsal horn neurons
between sympathetic fibers and afferent fibers has not been include: changes in neurotransmitters and tachykinins and
experimentally validated so far (51). In addition to this in their receptors (56-59), aberrant remodeling of peripheral
chemical coupling mechanism, aberrant structural changes afferent fibers and the synaptic dendrites in the SDH (60,61),
can also occur between sympathetic and afferent neurons. and the loss of inhibitory GABAergic inputs (62,63).
Under healthy conditions, most sympathetic postganglionic
fibers project distally to peripheral target cells via gray rami
Central mechanisms on the spinal cord
and a small quantity of fibers project proximally and usually
target along blood vessels. After nerve injury, however, Neurotransmitters and tachykinins are released from the
many proximally projecting perivascular catecholamine- central prominence of the nociceptive neurons in the DRG.
containing axons start to sprout in the DRG and even form Glutamate is the main transmitter in the DRG neurons (57).
“baskets” along the cell bodies of injured nerve fibers (52). Tachykinins include SP and neurokinin and neurotension
The mechanisms underlying this collateral sprouting of (45,64). In addition, different kinds of receptors are also
postganglionic sympathetic fibers is partially due to the involved in the induction and maintenance of central
neurotrophic signals and their receptors enriched along sensitization. There are three kinds of glutamate receptors
the afferent cells with lesioned axons (50,53). Whether in the SDH: the ionotropic N-methyl-D-aspartate (NMDA)
such pathological changes in structure are related to pain receptor, non-NMDA receptors, and metabotropic
behavior in patients after peripheral nerve lesions is still glutamate receptors. When glutamate binds to non-
uncertain and further investigations are expected. NMDA receptors, the receptors are opened and sodium
influx depolarizes the neurons. However, under normal
conditions or during weak stimulation, the channels of
Central mechanisms of pain
NMDA receptors are closed because of the presence of
Although peripheral mechanisms play an important role in magnesium. When pathological pain occurs, the magnesium
the induction and transmission of nociceptive information, ion is removed by the tonic nociceptive stimulation. At
it alone cannot explain all the characteristics of pathological this time, the increased glutamate releasing can activate
pain. Pathological nociceptive inputs also induce central the NMDA receptor in the SDH, which causes large
sensitization. amounts of calcium flow into the neuron (65,66). Calcium
ions induce second-messenger cascades that increase
neuronal excitability. Thus, the NMDA receptors and their
Central sensitization
downstream molecules play an important role in nociceptive
The spinal cord is a pivotal gateway where the primary information transmission, central sensitization induction

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Xiangya Medicine, 2016 Page 7 of 10

and maintenance as well as the chronic pain deterioration. input in the SDH (63). The reduction of GABA release
Laboratorially and clinically, administration of the NMDA from the presynaptic terminals occurs during pathological
receptor antagonist can prevent central sensitization and pain, which finally induces a functional loss of GABAergic
inhibit pain responses of animals or patients (67). After inhibition to excitatory neurons in the SDH (73).
central sensitization occurs, the neuronal excitability and
sensitivity is increased and the response to nociceptive
Central mechanisms on supraspinal level
stimuli is exaggerated. This phenomenon is called “wind
up” (68,69). Recently, using an integrative research approaches in animal
Neuropeptides also play critical roles in the induction models, genetically manipulated mice or human patients,
and maintenance of central sensitization. SP and CGRP accumulated evidences have consistently suggested that
released from the central terminals of nociceptive neurons in chronic pain is due to long-term plastic changes along
the DRG can bind to receptors expressed on the neurons of sensory pathways. Plastic changes not only take place in
SDH and contribute to the persistent hyperalgesia. CGRP peripheral nociceptors and SDH but also in cortical areas
can facilitate glutamate release through the activation of and subcortical areas that are involved in the processing
CGRP receptors on terminals of primary afferent neurons of painful information. The region of the brain activated
(59). Additionally, SP activates neurokinin receptors which by noxious stimuli is termed the “pain matrix”, and is
couple to phospholipase C and several kinds of intracellular comprised of the primary/secondary somatosensory cortex,
messengers. After binding to receptors, SP’s downstream insular cortex, prefrontal cortex (PFC), anterior cingulate
effects include depolarizing the membrane and facilitating cortex (ACC), thalamus, limbic system, basal ganglia and
the function of α-amino-3-hydroxy-5-methyl-4-isoxazole- brainstem. Neuroimaging studies reveal neuronal activity
propionic acid receptor (AMPA) and NMDA receptors (59). in these regions during pathological pain (74). The ACC,
In addition, both CGRP and SP can activate transcription PFC, insular and amygdala are pain-related structures with
factors and then increase the expression of related genes a close morphological and functional interconnection. It
that contribute to long-term changes in the excitability is reported that synaptic plasticity occurs in these cortical
of SDH neurons and maintain hyperalgesia. The changes or subcortical areas, such as in the ACC and amygdala
abovementioned finally induce the plasticity of the SDH (75,76). In addition, neuronal activities in the ACC and
which is another important factor of central sensitization. IC are believed to be important for pain perception and
With augmented noxious inputs and prolonged activation unpleasantness. Cortical manipulations can modulate pain
of pain pathways, neural plasticity is occurs and remodeling behavior and pain-related memory traces. Thus, treating
of synapses in the SDH is induced. Type A-beta fibers, which chronic pain requires an understanding of plastic changes in
originally transmit peripheral proprioceptive information, somatosensory pathways.
can release SP and are involved in the nociceptive
inputs transmission under the pathological pain (70).
Acknowledgements
More than 70% excitatory synapses are located on the
dendritic spine in the CNS, specifically on pyramidal cells None.
and neurons in the SDH. Dendritic spines can receive
highly convergent inputs from different origination (71),
Footnote
and then regulate synaptic transmission (72). After disease
and injury, such as the pathological pain discussed here, the Conflicts of Interest: This article has been originally published
dendritic spine morphology can change. Mushroom shape in the book The Art and Science of Palliative Medicine.
dendritic spines are increased, which indicates the increased
synaptic efficacy. New dendritic spines grow from the
References
dendrites of neurons, which can provide more postsynaptic
binding sites (71). All the changes of dendritic spines 1. Greene SA. Chronic pain: pathophysiology and treatment
contribute to the hyperexcitibility and central sensitization implications. Top Companion Anim Med 2010;25:5-9.
of spinal neurons. 2. Katz WA, Rothenberg R. Section 3: the nature of pain:
The third mechanism underlying the central pathophysiology. J Clin Rheumatol 2005;11:S11-5.
sensitization is the disinhibitatory action of GABAergic 3. Barkin RL, Barkin D. Pharmacologic management of

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Page 8 of 10 Xiangya Medicine, 2016

acute and chronic pain: focus on drug interactions and arousal by cholinergic and orexinergic systems. Ann N Y
patient-specific pharmacotherapeutic selection. South Med Acad Sci 2008;1129:26-34.
J 2001;94:756-70. 22. Bosshard SC, Baltes C, Wyss MT, et al. Assessment
4. Schaible HG, Richter F. Pathophysiology of pain. of brain responses to innocuous and noxious electrical
Langenbecks Arch Surg 2004;389:237-43. forepaw stimulation in mice using BOLD fMRI. Pain
5. Chapman CR, Gavrin J. Suffering: the contributions of 2010;151:655-63.
persistent pain. Lancet 1999;353:2233-7. 23. Casey KL, Lorenz J, Minoshima S. Insights into the
6. Twycross R, Harcourt J. A survey of pain in patients with pathophysiology of neuropathic pain through functional
advanced cancer. J Pain Symptom Manage 1996;12:273-82. brain imaging. Exp Neurol 2003;184:S80-8.
7. Mercadante S. Malignant bone pain: pathophysiology and 24. Navarro X, Vivó M, Valero-Cabré A. Neural plasticity
treatment. Pain 1997;69:1-18. after peripheral nerve injury and regeneration. Prog
8. Maxwell K. The challenges of cancer pain assessment and Neurobiol 2007;82:163-201.
management. Ulster Med J 2012;81:100-1. 25. Saadé NE, Jabbur SJ. Nociceptive behavior in animal
9. Dworkin RH, Backonja M. Advances in neuropathic pain: models for peripheral neuropathy: spinal and supraspinal
diagnosis, mechanisms, and treatment recommendations. mechanisms. Prog Neurobiol 2008;86:22-47.
Arch Neurol 2003;60:1524-34. 26. Newman PP. Visceral afferent functions of the nervous
10. Pasero C. Pathophysiology of neuropathic pain. Pain system. Monogr Physiol Soc 1974;(25):1-273.
Manag Nurs 2004;5:3-8. 27. Wu SX, Wang W, Li H, et al. The synaptic connectivity
11. Sommer C, Birklein F. Resolvins and inflammatory pain. that underlies the noxious transmission and modulation
F1000 Med Rep 2011;3:19. within the superficial dorsal horn of the spinal cord. Prog
12. Armstrong D, Dry RM. Observations on chemical excitants Neurobiol 2010;91:38-54.
of cutaneous pain in man. J Physiol 1953;120:326-51. 28. Kumazawa T, Perl ER. Excitation of marginal and
13. Purves D, Augustine GJ, Fitzpatrick D, et al., editors. substantia gelatinosa neurons in the primate spinal
Neuroscience. 2nd edition. Sunderland (MA): Sinauer cord: indications of their place in dorsal horn functional
Associates, 2001. organization. J Comp Neurol 1978;177:417-34.
14. Bessou P, Perl ER. Response of cutaneous sensory 29. Sugiura Y, Lee CL, Perl ER. Central projections of
units with unmyelinated fibers to noxious stimuli. J identified, unmyelinated (C) afferent fibers innervating
Neurophysiol 1969;32:1025-43. mammalian skin. Science 1986;234:358-61.
15. Djouhri L, Lawson SN. Abeta-fiber nociceptive primary 30. Kwiat GC, Basbaum AI. The origin of brainstem
afferent neurons: a review of incidence and properties in noradrenergic and serotonergic projections to the
relation to other afferent A-fiber neurons in mammals. spinal cord dorsal horn in the rat. Somatosens Mot Res
Brain Res Brain Res Rev 2004;46:131-45. 1992;9:157-73.
16. Snider WD, McMahon SB. Tackling pain at the source: 31. Fields HL, Basbaum AI. Central nervous system
new ideas about nociceptors. Neuron 1998;20:629-32. mechanisms of pain modulation. In: Wall PD, Melzack R,
17. Bester H, Matsumoto N, Besson JM, et al. Further editors. Textbook of Pain. London: Churchil Livingstone,
evidence for the involvement of the spinoparabrachial 1999:309-29.
pathway in nociceptive processes: a c-Fos study in the rat. 32. Fields HL, Vanegas H, Hentall ID, et al. Evidence that
J Comp Neurol 1997;383:439-58. disinhibition of brain stem neurones contributes to
18. Fields HL, Anderson SD, Wagner GM. The spinoreticular morphine analgesia. Nature 1983;306:684-6.
tract: an alternate pathway mediating pain. Trans Am 33. Fields HL, Heinricher MM, Mason P. Neurotransmitters
Neurol Assoc 1974;99:211-3. in nociceptive modulatory circuits. Annu Rev Neurosci
19. Foreman RD, Blair RW, Weber RN. Viscerosomatic 1991;14:219-45.
convergence onto T2-T4 spinoreticular, spinoreticular- 34. Zhang L, Sykes KT, Buhler AV, et al. Electrophysiological
spinothalamic, and spinothalamic tract neurons in the cat. heterogeneity of spinally projecting serotonergic and
Exp Neurol 1984;85:597-619. nonserotonergic neurons in the rostral ventromedial
20. Schott GD. Delayed onset and resolution of pain: some medulla. J Neurophysiol 2006;95:1853-63.
observations and implications. Brain 2001;124:1067-76. 35. Antal M, Petko M, Polgar E, et al. Direct evidence of
21. Jones BE. Modulation of cortical activation and behavioral an extensive GABAergic innervation of the spinal dorsal

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Xiangya Medicine, 2016 Page 9 of 10

horn by fibres descending from the rostral ventromedial 51. Chen Y, Michaelis M, Janig W, et al. Adrenoreceptor
medulla. Neuroscience 1996;73:509-18. subtype mediating sympathetic-sensory coupling in injured
36. Kato G, Yasaka T, Katafuchi T, et al. Direct GABAergic sensory neurons. J Neurophysiol 1996;76:3721-30.
and glycinergic inhibition of the substantia gelatinosa from 52. McLachlan EM, Janig W, Devor M, et al. Peripheral nerve
the rostral ventromedial medulla revealed by in vivo patch- injury triggers noradrenergic sprouting within dorsal root
clamp analysis in rats. J Neurosci 2006;26:1787-94. ganglia. Nature 1993;363:543-6.
37. Millan MJ. Descending control of pain. Prog Neurobiol 53. Deng YS, Zhong JH, Zhou XF. Effects of endogenous
2002;66:355-474. neurotrophins on sympathetic sprouting in the dorsal root
38. Ringkamp M, Meyer RA. Physiology of nociceptors. In: ganglia and allodynia following spinal nerve injury. Exp
Basbaum AL, Bushnell MC, editors. Science of pain. San Neurol 2000;164:344-50.
Diego: Academic Press, 2009:97-114. 54. Fornasari D. Pain mechanisms in patients with chronic
39. Michaelis M, Habler HJ, Jaenig W. Silent afferents: a pain.Clin Drug Investig 2012;32 Suppl 1:45-52.
separate class of primary afferents? Clin Exp Pharmacol 55. Ji RR, Kohno T. Central sensitization and LTP: do pain
Physiol 1996;23:99-105. and memory share similar mechanisms? Trends Neurosci
40. Schaible HG, Ebersberger A, Natura G. Update on 2003;26:696-705.
peripheral mechanisms of pain: beyond prostaglandins and 56. Zimmermann M. Pathobiology of neuropathic pain. Eur J
cytokines. Arthritis Res Ther 2011;13:210. Pharmacol 2001;429:23-37.
41. Aubdool AA, Brain SD. Neurovascular aspects of skin 57. Fundytus ME. Glutamate receptors and nociception:
neurogenic inflammation. J Investig Dermatol Symp Proc implications for the drug treatment of pain. CNS Drugs
2011;15:33-9. 2001;15:29-58.
42. Devor M. Ectopic generator. In: Basbaum AL, Bushnell 58. Harden RN. Chronic neuropathic pain. Mechanisms,
MC, editors. Science of pain. San Diego: Academic Press, diagnosis, and treatment. Neurologist 2005;11:111-22.
2009:83-7. 59. Seybold VS. The role of peptides in central sensitization.
43. Shim B, Kim DW, Kim BH, et al. Mechanical and Handb Exp Pharmacol 2009;(194):451-91.
heat sensitization of cutaneous nociceptors in rats with 60. Tan AM, Chang YW. Rac1-regulated dendritic spine
experimental peripheral neuropathy. Neuroscience remodeling contributes to neuropathic pain after
2005;132:193-201. peripheral nerve injury. Exp Neurol 2011;232:222-33.
44. Devor M. Sodium channels and mechanisms of 61. Tan AM, Waxman SG. Spinal cord injury, dendritic spine
neuropathic pain. J Pain 2006;7:S3-12. remodeling, and spinal memory mechanisms. Exp Neurol
45. Beydoun A, Backonja MM. Mechanistic stratification 2012;235:142-51.
of antineuralgic agents. J Pain Symptom Manage 62. Bridges D, Thompson SW, Rice AS. Mechanisms of
2003;25:S18-30. neuropathic pain. Br J Anaesth 2001;87:12-26.
46. Bossut DF, Perl ER. Effects of nerve injury on 63. Drew GM, Siddall PJ. Mechanical allodynia following
sympathetic excitation of A delta mechanical nociceptors. J contusion injury of the rat spinal cord is associated with
Neurophysiol 1995;73:1721-3. loss of GABAergic inhibition in the dorsal horn. Pain
47. Nickel FT, Seifert F, Lanz S, et al. Mechanisms 2004;109:379-88.
of neuropathic pain. Eur Neuropsychopharmacol 64. Basbaum AI. Spinal mechanisms of acute and persistent
2012;22:81-91. pain. Reg Anesth Pain Med 1999;24:59-67.
48. Xie Y, Zhang J, Petersen M. Functional changes in dorsal 65. Gordon DB, Love G. Pharmacologic management of
root ganglion cells after chronic nerve constriction in the neuropathic pain. Pain Manag Nurs 2004;5:19-33.
rat. J Neurophysiol 1995;73:1811-20. 66. Siddall PJ, Cousins MJ. Spinal pain mechanisms. Spine
49. Ali Z, Raja SN, Wesselmann U, et al. Intradermal 1997;22:98-104.
injection of norepinephrine evokes pain in patients with 67. O’Connor AB, Dworkin RH. Treatment of neuropathic
sympathetically maintained pain. Pain 2000;88:161-8. pain: an overview of recent guidelines. Am J Med
50. Drummond PD. Sensory disturbances in complex 2009;122:S22-32.
regional pain syndrome: clinical observations, autonomic 68. Davies SN, Lodge D. Evidence for involvement of
interactions, and possible mechanisms. Pain Med N-methylaspartate receptors in ‘wind-up’ of class
2010;11:1257-66. 2 neurones in the dorsal horn of the rat. Brain Res

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2


Page 10 of 10 Xiangya Medicine, 2016

1987;424:402-6. functions in spinal dorsal horn. Neuropharmacology


69. Herrero JF, Laird JM. Wind-up of spinal cord neurones 2011;60:921-9.
and pain sensation: much ado about something? Prog 74. Hirano S. Neuroimaging findings of central nervous
Neurobiol 2000;61:169-203. system dysfunction in neuropathic pain. Brain Nerve
70. Gracely RH. Pain measurement. Acta Anaesthesiol Scand 2012;64:1267-72.
1999;43:897-908. 75. Han JS, Neugebauer V. mGluR1 and mGluR5 antagonists
71. Yuste R, Majewska A. On the function of dendritic spines. in the amygdala inhibit different components of audible
Neuroscientist 2001;7:387-95. and ultrasonic vocalizations in a model of arthritic pain.
72. Bourne J, Harris KM. Do thin spines learn to be Pain 2005;113:211-22.
mushroom spines that remember? Curr Opin Neurobiol 76. Zhao MG, Toyoda H. Roles of NMDA NR2B subtype
2007;17:381-6. receptor in prefrontal long-term potentiation and
73. Cao J, Yang X. GABAergic disinhibition induced pain contextual fear memory. Neuron 2005;47:859-72.
hypersensitivity by upregulating NMDA receptor

doi: 10.21037/xym.2016.06.05
Cite this article as: Wu SX, Wang YY, Shi J, Dong YL, Cai GH.
Pathophysiology of pain. Xiangya Med 2016;1:2.

© Xiangya Medicine. All rights reserved. xym.amegroups.com Xiangya Med 2016;1:2

You might also like