Parkinsonism and Related Disorders: Review

You might also like

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

Parkinsonism and Related Disorders 19 (2013) 285e294

Contents lists available at SciVerse ScienceDirect

Parkinsonism and Related Disorders


journal homepage: www.elsevier.com/locate/parkreldis

Editor’s comment: Non motor symptoms are increasingly recognized to be of clinical importance for Parkinson’s disease (PD). Pain is
a universal symptom associated with almost every medical condition. Pain in PD is complex as the symptom can be multifactorial in origin.
In this article, Fil and colleagues conducted a detailed literature review of published literature on the possible mechanisms, classifications,
evaluation and potential risk factors of pain in PD. In the review, the authors highlight the limitations of published studies and the diffi-
culties in drawing guidelines and conclusions as there are significant methodological differences among published studies. This important
review draws our attention to the need for a concerted effort to conduct a multicenter evaluation with similar standardized protocols so
that possible mechanisms for pain in PD could be better understood, and risk factors could be identified early to enable more optimized
management of this disabling problem.
Eng-King Tan, Associate Editor, Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Outram road,
Singapore
Review

Pain in Parkinson disease: A review of the literature


[Universally Available]
Ayla Fil a, Roberto Cano-de-la-Cuerda b, Elena Muñoz-Hellín b, Lidia Vela c, María Ramiro-González d,
César Fernández-de-las-Peñas b, *
a
Neurologic Rehabilitation Unit, Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey
b
Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Universidad Rey Juan Carlos, Alcorcón, Madrid, Spain
c
Department of Neurology, Division of Movements Disorders, Hospital Universitario Fundación Alcorcón (HUFA), Alcorcón, Madrid, Spain
d
Emergency Department, Hospital de Madrid, Madrid, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Parkinson’s disease (PD) is a degenerative neurological disease presenting with motor and non-motor
Received 1 August 2012 signs and symptoms. Approximately 30e50% of the patients experience pain. There is no consensus
Received in revised form regarding the mechanisms and classification of pain in PD. This paper reviews current data on the
7 November 2012
possible mechanisms, classifications, evaluation and potential risk factors for pain in PD. Literature
Accepted 22 November 2012
searches were performed to identify clinical trials and reviews covering patho-physiology, classification,
type, evaluation and risk factors associated with pain in PD. Pain in PD could be related to pathologic
Keywords:
changes in the anatomic structures involved in nociceptive mechanisms. Studies on pain mechanisms
Classification
Evaluation
have been mostly conducted in animals. The mechanism of pain is complicated and influenced by
Pain different factors. There are several methodological differences between the studies trying to classify pain
Parkinson’s disease and to characterize its subtypes. Potential risk factors for pain in PD include: age, gender, and duration of
Risk factors the disease. Although pain is one of the non-motor symptoms most frequency experienced by patients, it
Review is often under recognized and inadequately treated in contrast to motor symptoms Multicenter studies
are needed that include a large cohort of subjects evaluated in multiple dimensions including pain in
order to obtain more data and to allow improved management of pain in patients with PD.
Ó 2012 Elsevier Ltd. Open access under CC BY-NC-ND license.

1. Introduction pathways [1]. Its prevalence in the general population is 0.1e0.3%


[2], showing an increase in individuals aged  65 years [3].
Parkinson’s disease (PD) is a chronic, progressive neurodegen- Cardinal findings in PD are tremor, rigidity, akinesia (i.e., bradyki-
erative disease characterized by loss of nigrostriatal dopaminergic nesia, hypokinesia) and postural instability. In addition to motor

* Corresponding author. Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Avenida de Atenas s/n, 28922 Alcorcón, Madrid, Spain. Tel.: þ34 91 488 88 84;
fax: þ34 91 488 89 57.
E-mail addresses: cesarfdlp@yahoo.es, cesar.fernandez@urjc.es (C. Fernández-de-las-Peñas).

1353-8020 Ó 2012 Elsevier Ltd. Open access under CC BY-NC-ND license.


http://dx.doi.org/10.1016/j.parkreldis.2012.11.009
286 A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294

disturbances, non-motor signs and symptoms are also common in increase their activity. The result is a reduction in the capacity of t-
these patients. These symptoms are classified as autonomic, i.e., cells to receive the stimuli or react to them. This is, essentially, the
hyperhidrosis, orthostatic hypotension, sexual-urinary dysfunction, gate-theory mechanism at the spinal level [12].
thermoregulation changes, cardiovascular disturbances, peripheral Two phylogenetically distinct systems, the medial and lateral
edema, dilated pupillae), sleep disturbances, neuropsychiatric pain systems, transmit pain to higher center brain neurons. The
problems, i.e., apathy, fatigue, anhedonia, depression, anxiety, medial system is mainly constituted of paleospinothalamic, spi-
panic attacks, dementia, psychosis, and sensory, i.e., internal nomesencephalic, spinoreticular, spinoparabrachial hypothalamic
tremor, restless leg syndrome, numbness, paresthesia, visual and spinothalamic tract fibers. These fibers travel in a caudal and
disturbances, and pain [4e7]. Among these sensory symptoms, rostral direction to higher centers by terminating in the para-
pain is observed in approximately 30e50% of PD patients; however, brachial nucleus, the locus cæruleus (reticular formation), the peri-
the incidence can increase to 68e85% when all types of pain are aqueductal gray substance (mesencephalon), intra-laminar and
taken into account [8]. Pain can appear at any time during the medial thalamic nuclei, thalamic ventral caudal parvocellular
disease, and can be present before diagnosis [9]. There is no nucleus and ventral caudal portae, the insula, parietal operculum,
consensus on the classification and the mechanisms of pain in PD the secondary somatosensory cortex, the amygdale and hippo-
patients. The objective of this review is to review the available data campus (Fig. 1). The medial system is involved in the affective and
on the possible mechanisms, classification, evaluation and poten- cognitive-evaluative dimension of pain, pain memory, and auto-
tial risk factors for pain in individuals with PD. nomic responses. Similarly, the lateral system is formed by the
neospinothalamic, the neotrigeminothalamic, and the cervical
2. Methods bundle and the beam of the dorsal horn. In the higher centers, these
fibers terminate in the lateral thalamus, the primary and secondary
2.1. Data sources
somatosensory areas, the parietal operculum and the insula (Fig. 1).
We performed computerized English language literature searches to identify The lateral system is important for the sensory-discriminative
clinical and controlled trials and reviews of pain in patients with PD making use of component of pain since it provides information about pain local-
the following databases: PubMed (from 1975), Ovid MEDLINE (from 1975), Ovid ization and duration [10,11,13,14].
EMBASE (from 1975), Cochrane Database of Systematic Reviews (CDSR), Cochrane
Collaboration Trials Register (CCTR), Cumulative Index to Nursing and Allied Health
The descending pathways originating in the brain stem and
Literature (CINAHL) and PEDro (Physiotherapy Evidence Database). cerebral structures also play an important role in the integration
We used the following Medical Subject Headings (MeSH) and main key words and modulation of nociceptive information in the dorsal horn. The
for searches: classification, patho-physiology, assessment, risk factors, Parkinson serotonergic, noradrenergic and dopaminergic networks are the
disease or pain. The search was supervised by a librarian scientist who helped us
principal components of these descending pain mechanisms. The
during each stage of the search. When database facilities allowed search limits,
searches were restricted to clinical or controlled trials. We also reviewed the sensitivity of the dorsal horn neurons can be increased or decreased
reference lists of the papers that we identified during the searches. Two authors by these pathways [11].
independently reviewed the selected articles. Published proceedings and abstracts PD, as a multifocal degenerative and progressive disease, could
were excluded. affect the pain process at multiple levels, from the transmission of
the pain from peripheral structures to the higher centers, to its
2.2. Data-extraction
reception and interpretation as well as interfering with several
As with article selection, data from each study were extracted independently by anatomic structures involved in pain mechanism. In a study with
2 authors. A standardized data-extraction form containing questions on population, PD patients, Nolano et al. showed that significant losses occur at the
methodology, results, and outcome measures was used according to the Consoli- level of free nerve endings and encapsulated nerve endings (i.e.,
dated Standards of Reporting Trials (CONSORT) statement. For each selected article,
Meissner’s corpuscles), independently of age or disease duration
the following data were recorded: inclusioneexclusion criteria, design, description
of study subject attrition and subject/investigator blinding, outcome measures, and [15]. They indicated that these changes in receptor size and
results. Finally, both authors had to achieve a consensus on each item on the data- peripheral deafferentation could play a relevant role in the patho-
extraction form. In case of no consensus, a third author helps in the decision. genesis of the sensory dysfunction of PD [15]. Starting in early stage
PD, degenerative changes can also occur in the spinal cord. Certain
3. Results neuronal losses have been observed in Lamina I of the posterior
spinal horn [16].
3.1. Patho-physiology of pain and Parkinson’s disease Braak et al. divided the disease into 6 periods [17,18]. The
changes typical of the pre-motor period start in the olfactory bulb
In 1986, pain was defined as a sensory and emotional experience and progress toward the inferior brain stem area (including the
associated with real or potential injuries or described in terms of medulla oblongata and pontine tegmentum) of Lewy neurites and
such injuries, by the International Association for the Study of Pain Lewy bodies (period 1e2). In the following symptomatic periods,
(IASP) [10]. It is known that several anatomic structures are pathologic changes take place in the mid brain including the sub-
involved at the same time in nociception. The process leading to stantia nigra (period 3), the meso-cortex (period 4), and finally the
pain starts with the stimulation of nociceptors. The thinly myelin- neo-cortex (periods 5e6) [17,18]. Nociceptive information cannot
ated Ad nociceptors response to mechanical and thermal stimuli, be transmitted directly from the spinal cord to higher centers
while the un-myelinated C-fiber nociceptors (polymodal) usually [19,20], since it is modulated by the descending pathways involving
respond to mechanical, thermal, or chemical stimulation. The various brain stem nuclei. Some of these nuclei are affected early in
stimuli from nociceptors arrive to the dorsal horn neurons of the PD [21]. As a result, this classification into 6 periods can be helpful
spinal cord. The lamina II, also known as substantia gelatinosa, plays for understanding those changes taking place in the anatomical
a major role in pain modulation at the spinal cord [10,11]. This area structures of the pain related higher centers during the course
forms an intermediate system regulating the transmission to the t- of PD.
cells on the lamina V, which in turn mediates the transmission of When examining the brain stem in PD, it is observed that ros-
sensory stimuli to the brain. The substantia gelatinosa system acts as tromedial medulla, comprising the nucleus raphe magnus and
an inhibitory mechanism on the t-cells. Stimulation of the Ad and C gigantocellular reticular nucleus, starts to be affected in period 2 of
fibers inhibits the substantia gelatinosa cells, reducing the output PD. This area is important for its role in the descending regulation
and their inhibitory action on the t-cells. As a result, the t-cells of pain since it is the last station of the descending anti-nociceptive
A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294 287

Fig. 1. Central pain pathways in Parkinson’s disease. Left: The lateral system is formed by the neospinothalamic, the neotrigeminothalamic, the cervical bundle and the beam of the
dorsal horn which fibers terminate in the lateral thalamus, the primary and secondary somatosensory areas, the parietal operculum and the insula. Right: The medial system is
mainly constituted by the paleospinothalamic, spinomesencephalic, spinoreticular, spinoparabrachial hypothalamic and spinothalamic tract fibers which terminate in the para-
brachial nucleus, the locus cæruleus, the peri-aqueductal gray substance, intra-laminar and medial thalamic nuclei, thalamic ventral caudal parvocellular nucleus and ventral caudal
portae, the insula, parietal operculum, the secondary somatosensory cortex, the amygdale and hippocampus.

pathways [22,23]. The locus coeruleus plays a role in the nocicep- subthalamic nucleus, the internal globus pallidus, and the sub-
tive modulation within the thalamus [24]. Consequently, the stantia nigra pars reticulata. Hyperactivity in these areas leads to
presence of Lewy bodies in the coeruleus/sub-coeruleus area, a strong inhibition of the lateral thalamic region, which can affect
which also plays a role in the noradrenergic modulation of pain, as pain mechanisms in two different ways: 1), disinhibition of medial
well as in intra-laminar and medial thalamic nuclei, could have an spinothalamic-cortical pathways to the anterior cingulate cortex
effect on autonomous, motivational-emotional and cognitive- due to the disruption of the lateral spinothalamic-cortical pathway
evaluative responses [13e25]. Serotonergic input to the spinal to the parietoinsular system [30]; 2), inhibition of the lateral thal-
dorsal horn from the caudal raphe affects the mechanisms of pain amus reducing localization, one of the sensory discrimination
by facilitating the nociceptive transmission of the mechanical elements of pain. The difficulty that patients have in localizing their
stimuli, and through such facilitation repressing the first reaction to pain symptoms supports this hypothesis. On the other hand, other
neurologic inflammation and to the chemical activation of primary properties of sensory discrimination become more sensitive to
afferents [26]. The caudal raphe nuclei and the nucleus raphe sensory stimuli [13]. This may explain the changes in pressure and
magnus, which are reciprocally linked [27], together with the thermal pain thresholds observed in PD.
coeruleus/sub-coeruleus area, make up the functional gain-setting Animal studies analyzing the effects of basal ganglia and
system. This system transforms important inputs from the higher- dopamine on sensory pathways and their contribution to sensory
level components of the limbic lobe into descending projections by perception reflect the importance of this region for pain [19].
controlling the excitability of the spinal and medullary pre-motor The neurons within the substantia nigra are responsive to low-
and motor neurons of the somato-motor system [28,29]. intensity deep mechanical stimuli (e.g. brushing, tapping, pressure)
Together with the locus coeruleus, the gigantocellular nucleus and electrical stimuli which decrease or increase the discharge ratio
and the nuclei of the bulbar raphe, the peri-aqueductal gray matter as compared to resting discharge. The nigral neuron responses can
and the parabrachial nuclei play an important role in the modula- depend on behavioral situations including the application of low-
tion of spinal nociceptive transmission, e.g., on inhibition of noci- intensity stimuli. As with non-nociceptive nigral neurons, noci-
ceptive stimuli coming from the dorsal horn neuron. A disturbance ceptive neurons within the substantia nigra may respond with
in this pain-inhibiting region could cause an increase in the suppression or enhancement of discharge frequency following
sensation of pain [13]. noxious stimulation and have large receptive fields that often
The lateral thalamus, one of the higher centers important for include the whole body. The large receptive fields of nociceptive nigral
pain mechanisms, also plays a major role in central pain. Dener- neurons show that these neurons have no major effect on the spatial
vation of the nigrostriatal system increases neuronal activity in the localization of painful stimuli [19]. Some studies, however, have
288 A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294

reported the importance of these neurons on the codification of (i.e. originating in the nervous system) and secondary (other
noxious stimuli [31], indicating that certain nociceptive nigral neurons sources than the nervous system, e.g., the musculoskeletal) [47]. As
may play a relevant role in sensory discrimination [19]. Animal new knowledge has emerged regarding the mechanisms, frequency
experiments have shown that stimulating the substantia nigra leads to and other properties of pain, more comprehensive classifications
nociceptive inhibition by activating the spinal cord neurons through have been proposed (Table 1). Ford’s classification is the most
dopaminergic pathways [32e35]. Based on the results of these studies, commonly used [55]. Ford [55] and Goetz et al. [48] distinguish the
it could be postulated that involvement of the substantia nigra in following categories: musculoskeletal pain, radicular/neuropathic
patients with PD affects the discriminatory aspect of pain and pain, dystonia-related pain, akathitic discomfort/pain and central
behavioral pain responses. In addition, striatal neurons that respond to parkinsonian pain [48,55]. Finally, classifications that correlate the
low-threshold somatosensory stimulation also have large cutaneous pain, directly or indirectly, with the disease have recently been
receptive fields [36] and can play a role in the localization of and the proposed [48,55].
response to stimuli from circumscribed regions [37e39]. Musculoskeletal pain covers pain arising from muscular, joint
Multi-sensory inputs within the striatum lead to the view that this and postural etiologies, including muscle cramps. Muscle cramps or
area plays a role in the coordination of behavioral responses by tightness in patients with PD can affect any part of the body but it
integrating sensory information conveyed by different sensory appears more typically in the neck, arm, paraspinal or calf muscles;
methods. The wide dynamic range of neurons, which exhibit low while joint pain occurs most frequently in shoulder, hip, knee, and
mechanical thresholds, are sensitive to somatosensory stimuli across ankle. The prevalence of musculoskeletal pain ranges from 45% to
a broad range of stimulus intensities and are maximally responsive to 74% in those patients with PD experiencing pain [8,47,48,51e56].
noxious stimuli as well, some nociceptive neurons within the stria- This prevalence rate is much higher than that reported in the
tum could be coding for the intensity of noxious stimuli. The ability of general population (8%e25%) [47,51,52]. The treatment of muscu-
striatal neurons to receive nociceptive stimuli from a large receptive loskeletal pain in PD can vary depending on the cause. If the pain is
area might indicate that these structures do not play a role in the due primarily to parkinsonian rigidity, dopaminergic therapy,
spatial localization of painful stimuli; the striatum may, however, act physical therapy, and exercises are indicated [55].
in concert with somatotopically organized areas that are co-activated Radicular-neuropathic pain manifests itself with pain, numbness
and provide precise spatial information for the next level of integra- or weakness in the area of a nerve root as a consequence of nerve or
tion [19]. Dopamine secretion during painful stimuli has been iden- root compression [55]. Postural abnormalities and dystonia devel-
tified within the dorso-lateral striatum. This type of response depends oping during the disease can lead to this type of pain through
on the subjective perception of pain intensity. Additionally, the discopathy [57]. The prevalence of radicular-neuropathic pain in
ventral striatum is clearly related to the emotional dimension of the patients with PD experiencing pain ranges from 5% to 20%
human pain process and expectation [40]. In conclusion, striatum [8,48,51e54,56].
involvement in PD could affect the emotional dimension of pain and Dystonic pain is represented by dystonic spasms which are
the subjective perception of pain intensity. usually paroxysmal, spontaneous, or triggered by movement or
Dopamine may also modulate pain at different levels including activity. They can be observed in the extremities, the face and the
the spinal cord, thalamus, peri-aqueductal gray matter, the basal pharyngeal muscles and are considered among the most painful
ganglia and the cingulate gyrus [41]. Axons coming from key symptoms that a patient with PD may experience. The prevalence
dopaminergic areas, such as the substantia nigra pars compacta, of dystonia-related pain ranges from 8% to 47% [8,48,50e52,56] in
the ventral tegmental area and the hypothalamus project to mes- patients with PD experiencing pain. In contrast, this pain is not
ocortical, mesolimbic, nigrostriatal and tuberoinfundibular path- present in healthy people. Symptoms can be more intense as early
ways. These pathways mediate different non-motor symptoms, e.g., morning manifestation of dopaminergic deficiency, and their
cognition, sleep and pain [41]. Mendlin observed that serotonin was intensity decreases after dopaminergic medication [58]. In the old
not secreted after blocking postsynaptic D2 receptors in rats [42]. days with high dose levodopa patients had ON dystonia as well.
Serotonin secretion in the forebrain is dependent on intact local Central pain is an insistent pain not restricted to nerve or
dopaminergic neurotransmission, as serotonin has an important radicular territories that manifest as neuropathic sensory events
role in the sensory and emotional properties of pain. A lesion of the such as paresthesia or shooting pain. The patient usually describes
striatum dopamine terminals can also modify pain transmission it as a bizarre and unexplained painful sensation predominately in
[43]. A significant reduction of the nociceptive delay and an the more affected side and in the “OFF” state [47,59,60]. Central
acceleration of the initial and final periods of nociceptive stimula- pain can occur in different areas of the body including: the mouth,
tion have been observed with lesions of this area. This condition rectum, vagina, abdomen, chest and testes [61e64]. The prevalence
effectively increases the nociceptive stimuli [44]. Finally, in a posi- of central pain in patients with PD is 10%e12% [8,53]. Central pain
tron emission tomography (PET) study conducted by Brefel- can be mainly modified by dopaminergic medication as it seems to
Courbon et al. increased brain activity during “off” periods was be related to an abnormal function of the medial spinor-
observed in the right insular, right prefrontal and left anterior eticulothalamic pathways [55]. In cases refractory to standard
cingulate cortex when subjects were exposed to experimental pain treatment, conventional analgesics, opiates, tricyclics may be also
[20]. It was suggested that levodopa could reverse these results. In helpful [55].
several pain studies, changes in threshold were observed after the The term akathitic discomfort (akathisia) is used to describe
administration of levodopa [20,45,46]. These results reveal the subjective restlessness or the painful impulse to move continually.
importance of dopamine in pain transmission. It seems clear that this pain improves with levodopa treatment
[55].
3.2. Classification and types of pain in Parkinson’s disease (PD) Restless leg syndrome (RLS) is another source of pain in
patients with PD. It is defined as a sensory-motor disturbance
Pain in PD manifests in different parts of the body and different that is prominent at night and improves with movement. It
types of pain has been described. As a result, no consensus has been includes intense and disagreeable sensations (paresthesia and
reached on the classification of pain in PD. Classification generally dysesthesia) of the extremities, more pronounced in the lower
follows the etiology of the pain [8,47e54]. Snider et al. distin- extremities [65]. Some reports have indicated that the prevalence
guished two main groups of sensory and pain symptoms: primary of RLS in patients with PD ranges from 8% to 20% [66,67], which is
Table 1
Categorization and rate of pain in Parkinson’s disease.
Author Number subjects Pain evaluation Rate of pain Classification of pain Rate of pain types Localization Factors affecting pain
Snider et al. [47] 101 PD patients Medical record PD patients (40%) Pain related to central nervous PD patients/No PD Head or back, hand, foot Levodopa
149 No PD patients No PD (8%) system dysfunction 11/0 Restless leg syndrome
Burning 22/3
Tingling 21/1
Numbness 29/3
Pain
Goetz et al. [48] 95 PD patients McGill Questionnarie 19 female (44%) Muscular pain: muscle cramps, 32 (74%) Neck, paraspinal muscles, calf Fluctuations
(43 female, 52 male) 24 male (46%) tightness 12 (28%) muscles Levodopa
(43 pain, 52 no pain) Dystonic pain: painful dystonia 6 (14%) Feet Disability
Radicular and neuritic pain: tingling 1 (2%) All body Worsening of
Diffuse generalized pain: discomfot of 6 (14%) Shoulders, hips, knees, ankles Parkinsonian
akathisia 0 (0%) symptoms Posture
Joint pain: stiffness, pain with motion, Increased resting tone
confused with joint
Talamic pain: Constant, boring,
ineffable and poorly lokalized pain

A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294


Scott et al. [49] 943 PD patients Questionnarie: SP-1/SP-2 One or more types Pain reported by patients (there is or Female/Male Neck Gender
(356 female, 587 male) and areas of pain per not) 53.7%/44.9% Low back
patient Neck pain 48.3%/40.7% Calf muscle
Back pain 45.3%/41.4%
Muscle cramps
Giuffrida et al. [54] 388 PD patients UPDRS (motor) 269 (67%) Muscular pain: stiffness, cramps, 94% e Motor fluctuations
(202 male, 186 female) 40.6  19.6 One or more types and pseudo-cramps, pasms 51%
(269 pain, 119 no pain) 43.8  14.8 areas of pain per Osteo-ligamentar rheumatologic pain: 2%
49.4  14.9 patient articular, spinal, peri-articular
43.9  13.8 Neurogenic painful syndromes:
paresthesia, dysesthesia, burning
sensation
Tinazzi et al. [50] 117 PD patients VAS 47 (40%) Musculoskeletal pain: aching, 21% For all types of pain Motor complications
(50 male, 67 female) cramping, arthralgic, joint 8% Neck: 2.5%
(47 pain, 70 no pain) Dystonic pain: pain associated with 40% Shoulder: 4%
dystonic movements 4% Arm: 1%
Central primary pain: burning, tingling Back: 9%
formication, bizarre quality Leg or foot: 28%
Akathitic discomfort
Lee et al. [56] 123 PD patients Wisconsin Brief 85% (according to Nociceptive pain: somatic and visseral 94.7% Legs: 37.9% Not described
(64 female, 59 male) Pain Inventory, PACA) Neuropathic pain 5.3% Back: 16.5%
PACA and VAS Musculoskeletal pain 62.6% Feet: 11.2%
Dystonic pain 67% Arms: 10.5%
PD treatment-related pain 0.8% Neck: 9.1%
Other causes 4.1% Hands: 6.7%
Negre-Pages et al. [51] 450 PD patients VAS, DoPaMiP, 62% PD patients with Dystonic pain 36% Head: 8.7% Motor complications
(254 male, 196 female) McGill Pain Questionnaire, chronic pain Neuropathic pain 7% Back: 12.6% Depressive symptoms
98 control The Brief Pain Inventory 5.5% PD patients with Akathisia 5% Upper limbs: 21.4% Age
(53 male, 45 female) non-chronic pain Muscular pain: Deep aching, myalgia, 2% Lower limbs: 67% PD duration
58.2% control subject cramps, stiffness or articular discomfort 31% Head: 11% Dyskinesia
Non-PD pain Back: 61% Severity of PD
Upper limbs: 14% Levadopa
Lower limbs: 30%
Defazio et al. [52] 402 PD patients VAS PD Patients (70%) Dystonic pain 6.7% Neck or shoulder: 6.2% Depression
(148 female, 254 male) Healthy (63%) Muscular pain: cramping, spams, joint 25.4% Arm: 11% Medical conditions
317 matched controls pain, pain with motion 4.7% Back: 15.7% Age
(138 female, 179 male) Peripheral neuropathic: pain in the 4.5% Leg or foot: 19.9%
territory of a root or nerve
Central neuropathic pain: burning,
tingling, formication, or bizarre quality

289
(continued on next page)
290 A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294

much higher than the prevalence found in the general population

PD: Parkinson’s disease; UPDRS: Unified Parkinson’s Disease Rating Scale; VAS: Visual analogue Scale; PACA: Palliative Care Assessment Tool; DoPaMiP: Douleur et maladie de Parkinson en Midi-Pyrénées; LANSS: Leeds
Factors affecting pain

(1%) [66].
In conclusion, the prevalence of all types of pain experienced
Female gender

by individuals with PD is reported to be over 80% (Table 1);


however, discrepancies can be observed among the studies due to
Akathisia

different methodologies. Since pain and sensory symptoms in PD


represent a broad spectrum, a multidisciplinary approach to pain
management is recommended. Nevertheless, it has been found
that that 50% of patients with PD-related pain did not receive
treatment [8].

3.3. Pain assessment in Parkinson’s disease (PD)


Not described

Not described
Rate of pain types Localization

Pain thresholds have been evaluated with instruments such as


the visual analog scale or the McGill Pain Questionnaire in indi-
viduals with PD. Additionally, quantitative sensory testing
including thermal, electrical and mechanical pain thresholds have
been also assessed in patients with PD [15,20,45,46,59,68e72]
(Table 2). The results revealed several discrepancies including
patient’s profile, pain threshold assessment, part of the body
44.4%
11.1%
19.1%
12.7%
12.7%

examined, outcomes included, the assessment during ONeOFF


70%
20%
40%
10%

periods, and the use of levodopa. As a result, while some studies


had reported lower pain thresholds in individuals with PD
Mixt pain: more than one of type pain
Central parkinsonian pain: burning,

[45,46,69,72]; others report no differences [59,68,73], whereas one


study revealed higher thresholds [74]. Therefore, there is no
Radicular or neuropathic pain
Musculoskeletal pain: aching,

consensus on differences in pain thresholds between patients with


Radicular/neuropathic pain

cramping, arthralgic, joint

PD and controls due to the controversial nature of current findings


tingling, bizarre quality
Dystonia-related pain

resulting from differences in the studies’ methodologies. In addi-


Musculoskeletal pain
Classification of pain

tion, similar conclusions are reported in relation to the effect of


administration of L-dopa in sensory thresholds since some studies
Dystonic pain
Central pain

reported increased pain thresholds after the administration of L-


dopa, whereas others have reported no changes in pain thresholds
after L-dopa administration (Table 2).
Indeed, Dellapina et al. have demonstrated that treatment with
apomorphine did not also modify pain thresholds or pain-induced
cerebral activity in patients with PD when compared to placebo,
24% two type pain
53% one type pain

suggesting the possibility that monoamine systems other than


Rate of pain

dopaminergic systems are involved in pain modulation [75]. The


lack of an effect of apomorphine to treat PD-related pain suggests
Assessment of Neuropathic Symptoms and Signs; STAI-TX: Stait Trait Anxiety Inventory.

that dopamine agonists may not be as effective as L-dopa in the


VAS, LANSS, STAI-TX 1 and 2 65%

treatment of pain [75]. Therefore, future studies are required to


further determine the role of dopamine agonist drugs in the
management of pain in PD.
SF-36 Bodily Pain Scale,
Brief Pain Inventory

3.4. Factors potentially affecting pain in Parkinson’s disease (PD)


Pain evaluation

The presence of pain in PD is usually dependent of motor


problems e.g., rigidity, tremor, akinesia, or postural abnormalities.
In this section, we explore possible factor influencing the presence
of pain in PD.
(146 pain, 30 no pain)
(90 female, 86% male)

(42 female, 54 male)

3.4.1. Age
Number subjects
176 PD patients

96 PD patients

If we consider the mean age of this patient population, an


increase in the prevalence of musculoskeletal pain with joint
problems and degenerative changes elsewhere in the body would
seem to be expected. However, evidence surrounding this is
controversial. In the study by Goetz et al. patients with pain were
younger than those without pain [48]. Defazio et al. reported that
Hanagasi et al. [53]
Table 1 (continued )

dystonia-related pain was observed at earlier ages [52]. Nègre-


Beiske et al. [8]

Pagès et al. also observed that PD-related pain was seen at an earlier
age than pain unrelated to PD [51]. Studies reporting the absence of
correlation between age and pain have also been published
Author

[8,50,56]. It is important to note that age has not systematically


been considered in all studies and the correlation between the
Table 2
Pain assessment in Parkinson’s disease (PD).

Year Study design Assessment methods for pain threshold Body part evaluated Results
Djaldetti et al. [59] 2004 Group 1: 36 hemi-PD patients (13 female, 23 male, 62 Tactile thresholds by using von Frey Bilateral forearm Mechanical and warmth sensory thresholds
years old, disease duration 5.3, H&Yahr 1e2.5, UPDRS filaments equal in groups
III: 22.5  14.8, 21 with pain e VAS score was Warm sensation and heat pain thresholds Heat pain thresholds:
48.9  24.1) (HPT) PD patients HPT < control subject
Group 2: 15 patients with fluctions (65 years, 12 female, Evaluation: before taking their first PD patients with pain HPT < PD patients
3 male, disease duration 10.6, UPDRS III: 27.8  17.2, 12 daily medication for all group without pain
patients with pain - VAS score 55.7  21.43) Group 2: assessment in ON and OFF More affected side HPT < less affected side
Control group: 28 age-matched controls (58.9  6.6 period
years; 16 women and 12 men)
Brefel-Courbon et al. [20] 2005 Group: 9 PD patients without pain (6 male, 3 female, 65 Cold pain threshold (CPT) right hand by Right hand OFF: pain threshold in PD patients < controls
years, disease duration 9.6, Hoehn and Yahr 2e2.5, immersion in cold water (temperature: ON: pain threshold in PD patients ¼ controls
UPDRS II OFF: 25  9, on: 15  7) from 20  C to 0  C, reduced to 3  C by Levodopa response: pain threshold during OFF
Control group: 9 healthy controls (6 male, 3 female, 3  C) period in
age: 59 years) Hand temperature was measured by PD patients < pain threshold during ON period
using skin thermometer in OFF and ON
periods

A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294


Gerdelat mas et al. [45] 2007 Group: 13 PD patients without pain (4 female, 9 male, The nociceptive flexion reflex (RIII reflex) Right leg OFF: PD patients < healthy subjects
age: 62 years, disease duration 7.3 years, UPDRS ON: All subjects were evaluated in ON and ON: PD patients ¼ healthy subjects
10.8  8.8, UPDRS OFF: 21.2  11.8) OFF condition Levodopa response: pain threshold during OFF
Control group: 10 healthy controls (4 female, 6 male, period in
age: 58 years) PD patients < pain threshold during ON period
Vela et al. [68] 2007 Group: 50 PD patients (34 male, 16 female, age: 66 Pressure pain threshold (PPT) was The dorsal side of the proximal PPT was similar for three groups all body part
years, disease duration 8.0 / 25 patients without evaluated during ON period phalanx of the second finger Women had lower PPT than men
dyskinesia (UPDRS: 19.95  9.16, H&Y 2.28  0.68 and and brachioradialis muscle
25 patients with dyskinesia (UPDRS: 24.56  8.35, H&Y
1.88  0.50)
Control group: 25 controls (12 male and 13 female, 62
years)
Lim et al. [46] 2008 Group stable responders: 12 PD patients (8 male, 4 Cold pain threshold (CPT): first right Right and left hand CPT and tolerance (OFF) Healthy > PD patients
female, age 68 years, duration 3  1 years, UPDRS III hand and later left hand was immersed After levodopa administration dyskinetic
OFF: 30  3, UPDRS III ON: 26  3) in an ice-water bath (water patients exhibited a large increase in CPT and
Group fluctuators: 15 PD patients (7 male, 8 female, temperature 1.0e2.0  C) until subject tolerance that was absent in stable responders
age 64 years, duration 3  1 years, UPDRS III OFF: 27  3, felt “faintly painful”
UPDRS III ON: 21  3) To determinate “cold pain tolerance” the
Group dyskinetics: 23 PD patients (13 male, 10 female, hand was kept immersed until it
age 63 years, duration 6  1 years, UPDRS III OFF: 31  3, became ‘‘intolerable’’ or 180 s had
UPDRS III ON: 19  2) elapsed
Control group: 23 age and sex matched healthy Pain thresholds were evaluated in ON
subjects condition
Nolano et al. [15] 2008 Group: 18 PD patients (9 male, 9 female, age: 62.7 years, Tactile threshold: Semmes-Weinstein The dorsum of the hand and Tactile threshold: PD patients > control subjects
H&Y: 2.6  1.1, UPDRS III 26.6  10.2) monofilaments foot of the less affected side in Mechanical pain perception: PD
Control group: 30 age- and sex-matched healthy Mechanical pain perception: calibrated PD patients patients < control subjects
subjects (61 years old) monofilament, with a bending force of Thermal thresholds (cold, warm, cold pain and
95 mN heat pain):
Thermal thresholds (cold, warm, cold PD patients > control subjects
pain and heat pain thresholds)
All pain threshold tests were performed
during ON condition.
Mylius et al. [69] 2009 Group: 15 PD patients (8 patients with pain (PDP), 7 The nociceptive flexion reflex (NFR) Left or right calf NFR: PD patients < control subjects/PDN ¼ PDP
without pain (PDN), 6 female and 9 male, 63 years, Electrical pain threshold (EPT) patients
duration 11.0  5.4 years, UPDRS III 28.3  9.4, H&Y Heat pain thresholds (HPT) EPT: PD patients < controls PDN ¼ PDP patients
2.3  0.7) HPT: PD patients < control subjects/PDN ¼ PDP
Control group: 18 control subjects (11 female and 7 patients
male, 67 years old)
(continued on next page)

291
292 A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294

different kinds of pain with age has not been investigated in some

-Pain threshold: PD patients < control subjects

-Pain tolerance: PD patients < control subjects


studies.

subjects (hand) PD patients > controls (Feet)


-Tactile threshold: PD patients ¼ control 3.4.2. Gender
Current thinking tends to favor the absence of gender differ-
ences in relation to the presence or absence of pain [48,50e53];
however, Beiske et al. reported that female gender was a significant
PD OFF ¼ healthy controls
PD ON > healthy controls

predictor of pain in PD [8]. Scott et al. also observed that male and
female patients do not complain of pain in the same locations, since
women reported higher prevalence of neck and low back pain than
PD ON ¼ PD OFF

men [49]. Vela et al. [68] found lower pressure pain thresholds in
women. A link between pressure pain thresholds and musculo-
skeletal pain has been described [76], leading to the suggestion that
Results

(both)

(both)

mechanical sensitivity is higher in women.

3.4.3. Severity and duration of the disease


and to the big toe of each foot
(proximal, middle, and distal)
Volar surface of each forearm

The little finger of each hand

The effect of severity and duration of disease on pain is


controversial. Nègre-Pagès et al. observed a correlation between
PD-related pain and the severity and duration of disease [51].
Body part evaluated

Similarly, a longer duration of disease and greater motor compli-


cations in patients with PD with pain have been reported as
compared to pain-free patients [50,77]. Other studies have not
found this correlation [8,52,53].

3.4.4. Depression
Symptoms of depression can be observed in PD patients,
conditions and twice in healthy subjects

pain tolerance were measured by using


Assessment methods for pain threshold
VAS (pain) 1 e For pain: 0e200 mm: 0

Tactile threshold, pain threshold, and

ranging in intensity from mild to severe [78]. Considering that


e100 mm thermal sensation, 100

chronic pain is a risk factor in the development of depression in


HPT was assessed in ON and OFF

the elderly [79,80] and the older age and complications of PD


e200 mm painfull sensation

patients, a link between depression and pain seems probable.


Depression has been generally evaluated in studies of pain. Goetz
et al. reported that pain intensity correlated with depression, and
electrical stimuli.

that the latter is more severe in patients experiencing pain than


in those without pain, even though the incidence of depression
was the same in both groups of patients [81]. Starkstein et al.
observed that pain was more frequent in patients with major
depression as compared to those with milder depression [82].
Similarly, a link between pain and depression in PD has been
Group: 106 PD patients (66 with pain, 40 without pain,
Group: 12 PD patients (10 male, 2 female, age: 60 years,

Control group: 51 healthy control subjects (36 male, 15


Control group: 13 control subjects (7 male, 6 female,

reported by Ehrt et al. [77] and McNamara et al. [1]; however,


70 years, 59 male, 47 female, duration of disease: 5.7
duration 9.4  3.2, UPDRS ON 15  10.9, UPDRS OFF

years, H&Y: 2.1  0.9, UPDRS III OFF: 23.5  13.6)

others have not shown a correlation between pain and depres-


sion [50,53]. Current evidence suggests that depression is
a parameter to be considered in studies of pain in patients with
PD, considering the frequency of depression in PD and the fact
that it can modify the perception of pain and it can be also
properly treatable.

3.4.5. Systemic disease and pre-existing disturbances


Patients with pre-existing pain related to systemic diseases such
female, age: 69 years)
62.9  8.3 years old)

as diabetes, osteoporosis, rheumatic disease, and age-dependent,


e.g., joint or disc disease or postural problems, will continue to
Study design

experience these after diagnoses; however, new disease related


28.8  9.4

symptoms, such as rigidity, akinesia, muscular cramps, and postural


instability can worsen these difficulties. Beiske et al. reported that
some patients complained of pain before the diagnosis of PD [8].
The results reported by Defazio et al. showed that joint pain and
2011
2010
Year

peripheral neuropathic pain is seen in controls matched for age


with PD patients [52]. These studies support the theory that some
pain seen in patients with PD is related to age rather than the
Nandhagopal et al. [72]

disease itself.
Marsala et al. [71]
Table 2 (continued )

The pain experienced by the patients prior to PD diagnosis could


be one reason for the controversial character of the findings
regarding the incidence of pain in PD. Several studies had generally
evaluated the presence of pain in a cross-sectional design, without
indicating if the complaints started before or after the diagnosis of
PD. On the other hand, such pain could also be related to the
A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294 293

disease, but appearing much earlier than the diagnosis [83]. [6] Bayulkem K, Lopez G. Non-motor fluctuations in Parkinson’s disease: clinical
spectrum and classification. J Neurol Sci 2010;289:89e92.
Controlled studies in large patient populations defining the time at
[7] Wolters ECH. Non-motor extranigral signs and symptoms in Parkinson’s
which the pain started and investigating it in detail according to its disease. Parkinsonism Relat Disord 2009;3:S6e12.
different types are more than needed. [8] Beiske AG, Loge JH, Rønningen A, Svensson E. Pain in Parkinson’s disease:
prevalence and characteristics. Pain 2009;141:173e7.
[9] Tolosa E, Compta Y, Gaig C. The premotor phase of Parkinson’s disease.
3.5. Pain and quality of life Parkinsonism Relat Disord 2007;13:S2e7.
[10] Almeida TF, Roizenblatt S, Tufik S. Afferent pain pathways: a neuroanatomical
review. Brain Res 2004;1000:40e56.
Pain clearly affects the quality of life (Quol) of PD patients. A [11] Millan MJ. The induction of pain: an integrative review. Prog Neurobiol 1999;
link between pain, sleep problems and depression has been 57:1e164.
described [81,84]. The interactions among these conditions [12] Wall PD, Melzack R. Textbook of pain. Churchill London: Livingstone; 1994.
[13] Scherder E, Wolters E, Polman C, Sergeant J, Swaab D. Pain in Parkinson’s
increase and complicate the problem by creating a vicious circle. disease and multiple sclerosis: its relation to the medial and lateral pain
Some studies report that pain and other related conditions systems. Neurosci Biobehav Rev 2005;29:1047e56.
negatively impact Quol in PD patients [85,86]. Barone et al. found [14] Willis WD, Westlund KN. Neuroanatomy of the pain system and of the
pathways that modulate pain. J Clin Neurophysiol 1997;14:2e31.
that non-motor disturbances negatively influence Quol and that
[15] Nolano M, Provitera V, Estraneo A, Selim MM, Caporaso G, Stancanelli A, et al.
leg pain occupies third place in order of importance [87]. Politis Sensory deficit in Parkinson’s disease: evidence of a cutaneous denervation.
et al. examined the impact of PD from a patient’s perspective [88]. Brain 2008;131:1903e11.
[16] Braak H, Sastre M, Bohl JR, de Vos RA, Del Tredici K. Parkinson’s disease:
They classified patients into two groups according to the duration
lesions in dorsal horn layer I, involvement of parasympathetic and
of disease: early (<6 years) and late (>6 years), and asked them to sympathetic pre- and postganglionic neurons. Acta Neuropathol 2007;113:
determine their problems in order of importance. Pain was one of 421e9.
the most frequent non-motor problems in both groups. The early [17] Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of
brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging
PD patient group complained of musculo-cutaneous pain first and 2003;24:197e211.
visceral pain second, whereas the late PD group listed musculo- [18] Braak H, Rub U, Jansen-Steur EN, Del Tredici K, de Vos RA. Cognitive status
cutaneous pain in first place, but dyskinetic and dystonia-related correlates with neuropathologic stage in Parkinson disease. Neurology 2005;
64:1404e10.
pain second [88]. [19] Chudler EH, Dong WK. The role of the basal ganglia in nociception and pain.
Pain 1995;60:3e38.
[20] Brefel-Courbon C, Payoux P, Thalamas C, Ory F, Quelven I, Chollet F, et al.
4. Conclusions Effect of levodopa on pain threshold in Parkinson’s disease: a clinical and
positron emission tomography study. Mov Disord 2005;20:1557e63.
Pain in PD is likely related to pathologic changes in the [21] Millan MJ. Descending control of pain. Prog Neurobiol 2002;66:355e474.
[22] Kuraishi Y, Fukui K, Shiomi H, Akaike A, Takagi H. Microinjection of opioids
anatomical structures involved in nociceptive pain mechanisms.
into the nucleus reticularis gigantocellularis of the rat: analgesia and increase
Most studies on pain mechanism in PD have been conducted in in the normetanephrine level in the spinal cord. Biochem Pharmacol 1978;27:
animals. Pain, one of the most frequent non-motor symptoms 2756e8.
[23] Gebhart GF. Descending modulation of pain. Neurosci Biobehav Rev 2004;27:
affecting PD patients has complicated mechanism and is influenced
729e37.
by several factors, e.g., age, gender, depression, severity, or duration [24] Voisin DL, Guy N, Chalus M, Dallel R. Nociceptive stimulation activates locus
of the disease. Further, there are many methodological differences coeruleus neurones projecting to the somatosensory thalamus in the rat.
among the studies aimed at classifying pain and characterizing its J Physiol 2005;566:929e37.
[25] Pertovaara A. Noradrenergic pain modulation. Prog Neurobiol 2006;80:53e83.
subtypes. The fact that the patient population includes mostly the [26] Benarroch EE. Descending monoaminergic pain modulation: bidirectional
aged increases the number of factors involved. There are also control and clinical relevance. Neurology 2008;71:217e21.
problems with the studies investigating pain thresholds and [27] Clark FM, Proudfit HK. Projections of neurons in the ventromedial medulla to
pontine catecholamine cell groups involved in the modulation of nociception.
perception, as different studies follow different protocols and Brain Res 1991;540:105e15.
consequently have different outcomes. There is a need for multi- [28] Holstege G. The emotional motor system. Eur J Morphol 1992;30:67e79.
center studies with a large number of patients and controls, in [29] Nieuwenhuys R. The greater limbic system, the emotional motor system and
the brain. Prog Brain Res 1996;107:551e80.
which patients should be evaluated in different dimensions for [30] Craig AD. A new version of the thalamic disinhibition hypothesis of central
measurable endpoints to acquire definitive data and open new pain. Pain Forum 1998;7:1e14.
directions for treatment, of the pain so often present in patients [31] Gao DM, Jeaugey L, Pollak P, Benabid AL. Intensity-dependent nociceptive
responses from presumed dopaminergic neurons of the substantia nigra, pars
with PD.
compacta in the rat and their modification by lateral habenula inputs. Brain
Res 1990;529:315e9.
Conflict of interest [32] Barnes CD, Fung SJ, Adams WL. Inhibitory effects of substantia nigra on
impulse transmission from nociceptors. Pain 1979;6:207e15.
[33] Burkey AR, Carstens E, Jasmin L. Dopamine reuptake inhibition in the rostral
The authors declare no conflicts of interest. agranular insular cortex produces antinociception. J Neurosci 1999;19:4169e79.
1) No funds were received for this study; 2) No financial benefits [34] Li J, Ji YP, Qiao JT, Dafny N. Suppression of nociceptive responses in paraf-
ascicular neurons by stimulation of substantia nigra: an analysis of related
have or will be derived by the authors from this study; 3) The data inhibitory pathways. Brain Res 1992;591:109e15.
from this study have not been presented in any other form. [35] Baumeister AA, Anticich TG, Hawkins MF, Liter JC, Thibodeaux HF, Guillory EC.
Evidence that the substantia nigra is a component of the endogenous pain
suppression system in the rat. Brain Res 1988;447:116e21.
References [36] Sedgwick EM, Williams TD. The response of single units in the caudate
nucleus to peripheral stimulation. J Physiol 1967;189:281e98.
[1] McNamara P, Stavitsky K, Harris E, Szent-Imrey O, Durso R. Mood, side of [37] Carelli RM, West MO. Representation of the body by single neurons in the
motor symptom onset and pain complaints in Parkinson’s disease. Int J Geriatr dorsolateral striatum of the awake, unrestrained rat. J Comp Neurol 1991;309:
Psychiatry 2010;25:519e24. 231e324.
[2] Weintraub D, Comella CL, Horn S. Parkinson’s disease-Part 1: pathophysi- [38] Lidsky TI. Pallidal and entopeduncular single unit activity in cats during
ology, symptoms, burden, diagnosis, and assessment. Am J Manag Care 2008; drinking. Electroenceph Clin Neurophysiol 1975;39:79e84.
14:40e8. [39] Levine MS, Schneider JS, Lloyd RL, Hull CD, Buchwald NA. Aging reduces
[3] Alves G, Forsaa EB, Pedersen KF, Dreetz-Gjerstad M, Larsen JP. Epidemiology of somatosensory responsiveness of caudate neurons in the awake cat. Brain Res
Parkinson’s disease. J Neurol 2008;255:18e32. 1987;10(405):389e94.
[4] Park A, Stacy M. Non-motor symptoms in Parkinson’s disease. J Neurol 2009; [40] Juri C, Rodriguez-Oroz M, Obeso JA. The pathophysiological basis of sensory
256:293e8. disturbances in Parkinson’s disease. J Neurol Sci 2010;289:60e5.
[5] Simuni T, Sethi K. Nonmotor manifestations of Parkinson’s disease. Ann [41] Chaudhuri KR, Schapira AHV. Non-motor symptoms of Parkinson’s disease:
Neurol 2008;64:S65e80. dopaminergic pathophysiology and treatment. Lancet Neurol 2009;8:464e74.
294 A. Fil et al. / Parkinsonism and Related Disorders 19 (2013) 285e294

[42] Mendlin A, Martin FJ, Jacobs BL. Dopaminergic input is required for increases [66] Krishnan PR, Bhatia M, Behari M. Restless legs syndrome in Parkinson’s
in serotonin output produced by behavioral activation: an in vivo micro- disease: a case controlled study. Mov Disord 2002;18:181e5.
dialysis study in rat forebrain. Neuroscience 1999;93:897e905. [67] Ondo WG, Vuong KD, Jankovic J. Exploring the relationship between Parkin-
[43] Drake DF, Harkins S, Qutubuddin A. Pain in Parkinson’s disease: pathology to son disease and restless legs syndrome. Arch Neurol 2002;59:421e4.
treatment, medication to deep brain stimulation. NeuroRehabilitation 2005; [68] Vela L, Lyons KE, Lieberman AN. Pain-pressure threshold in patients with
20:335e41. Parkinson’s disease with and without dyskinesia. Parkinsonism Relat Disord
[44] Saad NE, Atweh SF, Bahuth NB, Jabbur SJ. Augmentation of nociceptive 2007;13:189e92.
reflexes and chronic deafferentation pain by chemical lesions of either [69] Mylius V, Engau I, Teepker M, Stiasny-Kolster K, Schepelmann K, Oertel WH.
dopaminergic terminals or midbrain dopaminergic neurons. Brain Res 1997; Pain sensitivity and descending inhibition of pain in Parkinson’s disease.
751:1e12. J Neurol Neurosurg Psychiatry 2009;80:24e8.
[45] Gerdelat-Mas A, Simonetta-Moreau M, Thalamas C, Ory-Magne F, Slaoui T, [70] Gierthmühlen J, Arning P, Binder A, Herzog J, Deuschl G, Wasner G, et al.
Rascol O, et al. Levodopa raises objective pain threshold in Parkinson’s Influence of deep brain stimulation and levodopa on sensory signs in Par-
disease: a RIII reflex study. J Neurol Neurosurg Psychiatry 2007;78:1140e2. kinson’s disease. Mov Disord 2010;25:1195e202.
[46] Lim SY, Farrell MJ, Gibson SJ, Helme RD, Lang AE, Evans AH. Do dyskinesia and [71] Marsala SZ, Tinazzi M, Vitaliani R, Recchia S, Fabris F, Marchini C. Spontaneous
pain share common pathophysiological mechanisms in Parkinson’s disease? pain, pain threshold, and pain tolerance in Parkinson’s disease. J Neurol 2011;
Mov Disord 2008;23:1689e95. 258:627e33.
[47] Snider SR, Fahn S, Isgreen WP, Cote LJ. Primary sensory symptoms in [72] Nandhagopal R, Troiano AR, Mak E, Schulzer M, Bushnell MC, Stoessl AJ.
parkinsonism. Neurology 1976;26:423e9. Response to heat pain stimulation in idiopathic Parkinson’s disease. Pain Med
[48] Goetz CG, Tanner CM, Levy M, Wilson RS, Garron DC. Pain in Parkinson’s 2010;11:834e40.
disease. Mov Disord 1986;1:45e9. [73] Massetani R, Lucchetti R, Vignocchi G, Siciliano G, Rossi B. Pain threshold and
[49] Scott B, Borgman A, Engler H, Johnels B, Aquilonius SM. Gender differences in polysynaptic components of the blink reflex in Parkinson’s disease. Funct
Parkinson’s disease symptom profile. Acta Neurol Scand 2000;102:37e43. Neurol 1989;4:199e202.
[50] Tinazzi M, Del Vesco C, Fincati E, Ottaviani S, Smania N, Moretto G, et al. Pain [74] Guieu R, Pouget J, Serratrice G. Seuil nociceptif et maladie de Parkinson. Rev
and motor complications in Parkinson’s disease. J Neurol Neurosurg Psychi- Neurol 1992;148:641e4.
atry 2006;77:822e5. [75] Dellapina E, Gerdelat-Mas A, Ory-Magne F, Purcel L, Galitzky M, Calvas F, et al.
[51] Nègre-Pagès L, Regragui W, Bouhassira D, Grandjean H, Rascol O, DoPaMiP Apomorphine effect on pain threshold in Parkinson’s disease: a clinical and
Study Group. Chronic pain in Parkinson’s disease: the cross-sectional French positron emission tomography study. Mov Disord 2011;26:153e7.
DoPaMiP survey. Mov Disord 2008;23:1361e9. [76] Rollman GB, Lautenbacher S. Sex differences in musculoskeletal pain. Clin J
[52] Defazio G, Berardelli A, Fabbrini G, Martino D, Fincati E, Fiaschi A. Pain as Pain 2001;17:20e4.
a nonmotor symptom of Parkinson disease: evidence from a case-control [77] Ehrt U, Larsen JP, Aarsland D. Pain and its relationship to depression in Par-
study. Arch Neurol 2008;65:1191e4. kinson disease. Am J Geriatr Psychiatry 2009;17:269e75.
[53] Hanagasi HA, Akat S, Gurvit H, Yazici J, Emre M. Pain is common in Parkinson’s [78] Tandberg E, Larsen JP, Aarsland D, Cummings JL. The occurrence of
disease. Clin Neurol Neurosurg 2011;113:11e3. depression in Parkinson’s disease. A community-based study. Arch Neurol
[54] Giuffrida R, Vingerhoets FJ, Bogousslavsky J, Ghika J. Pain in Parkinson’s 1996;53:175e9.
disease. Rev Neurol (Paris) 2005;161:407e18. [79] Cohen-Mansfield J, Marx MS. Pain and depression in the nursing home:
[55] Ford B. Pain in Parkinson’s disease. Mov Disord 2010;1:S98e103. corroborating results. J Gerontol 1993;48:96e7.
[56] Lee MA, Walker RW, Hildreth TJ, Prentice WM. A survey of pain in idiopathic [80] Parmelee PA, Katz IR, Lawton MP. The relation of pain to depression among
Parkinson’s disease. J Pain Symptom Manage 2006;32:462e9. institutionalized aged. J Gerontol 1991;46:15e21.
[57] Adams MA, Freeman BJC, Morrison HP, Nelson IW, Dolan P. Mechanical [81] Goetz CG, Wilson RS, Tanner CM, Garron DC. Relationships among pain,
initiation of intervertebral disc degeneration. Spine 2000;25:1625e36. depression, and sleep alterations in Parkinson’s disease. Adv Neurol 1987;45:
[58] Quinn NP. Classification of fluctuations in patients with Parkinson’s disease. 345e7.
Neurology 1998;51:S25e9. [82] Starkstein SE, Preziosi TJ, Robinson RG. Sleep disorders, pain and depression in
[59] Djaldetti R, Shifrin A, Rogowski Z, Sprecher E, Melamed E, Yarnitsky D. Parkinson’s disease. Eur Neurol 1991;31:352e5.
Quantitative measurement of pain sensation in patients with Parkinson [83] Stamey W, Davidson A, Jankovic J. Shoulder pain: a presenting symptom of
disease. Neurology 2004;62:2171e5. Parkinson disease. J Clin Rheumatol 2008;14:253e4.
[60] Schestatsky P, Kumru H, Valls-Solé J, Valldeoriola F, Marti MJ, Tolosa E, et al. [84] van Hilten JJ, Weggeman M, van der Velde EA, Kerkhof GA, van Dijk JG,
Neurophysiologic study of central pain in patients with Parkinson disease. Roos RA. Sleep, excessive daytime sleepiness and fatigue in Parkinson’s
Neurology 2007;69:2162e9. disease. J Neural Trans Park Dis Dement Sect 1993;5:235e44.
[61] Clifford TJ, Warsi MJ, Burnett CA. Burning mouth in Parkinson’ disease [85] Quittenbaum BH, Grahn B. Quality of life and pain in Parkinson’s disease:
sufferers. Gerodontology 1998;15:73e8. a controlled cross-sectional study. Parkinsonism Relat Disord 2004;10:
[62] Quigley EM. Gastrointestinal dysfunction in Parkinson’s disease. Semin Neurol 129e36.
1996;16:245e50. [86] Caap-Ahlgren M, Dehlin O. Insomnia and depressive symptoms in patients
[63] Ford B, Louis ED, Greene P, Fahn S. Oral and genital pain syndromes in Par- with Parkinson’s disease. Relationship to health-related quality of life. An
kinson’s disease. Mov Disord 1996;11:421e6. interview study of patients living at home. Arch Gerontol Geriatr 2001;32:
[64] Ku1akowska A, Pogorzelski R, Borowik H, Drozdowski W. Wearing off 23e33.
phenomenon presenting with features of paroxysmal abdominal pain. Neurol [87] Barone P, Antonini A, Colosimo C, Marconi R, Morgante L, Avarello TP. The
Neurochir Pol 2003;37:197e202. PRIAMO study: a multicenter assessment of non-motor symptoms and their
[65] Walters AS, the International Restless Legs Syndrome Study Group. impact on quality of life in Parkinson’s disease. Mov Disord 2009;24:1641e9.
Toward a better definition of the restless legs syndrome. Mov Disord [88] Politis M, Wu K, Molloy S, Bain P, Chaudhuri KR, Piccini P. Parkinson’s disease
1995;10:634e42. symptoms: the patient’s perspective. Mov Disord 2010;25:1646e51.

You might also like