Skip to main content
Erschienen in: The Journal of Headache and Pain 1/2017

Open Access 01.12.2017 | Research article

Calcitonin gene-related peptide and pain: a systematic review

verfasst von: Wendy Sophie Schou, Sait Ashina, Faisal Mohammad Amin, Peter J. Goadsby, Messoud Ashina

Erschienen in: The Journal of Headache and Pain | Ausgabe 1/2017

Abstract

Background

Calcitonin gene-related peptide (CGRP) is widely distributed in nociceptive pathways in human peripheral and central nervous system and its receptors are also expressed in pain pathways. CGRP is involved in migraine pathophysiology but its role in non-headache pain has not been clarified.

Methods

We performed a systematic literature search on PubMed, Embase and ClinicalTrials.gov for articles on CGRP and non-headache pain covering human studies including experimental studies and randomized clinical trials.

Results

The literature search identified 375 citations of which 50 contained relevant original data. An association between measured CGRP levels and somatic, visceral, neuropathic and inflammatory pain was found. In 13 out of 20 studies in somatic pain conditions, CGRP levels had a positive correlation with pain. Increased CGRP levels were reported in plasma, synovial and cerebrospinal fluid in subjects with musculoskeletal pain. A randomized clinical trial on monoclonal antibody, which selectively binds to and inhibits the activity of CGRP (galcanezumab) in patients with osteoarthritis knee pain, failed to demonstrate improvement of pain compared with placebo. No studies to date have investigated the efficacy of monoclonal antibodies against CGRP receptor in non-headache pain conditions.

Conclusion

The present review revealed the association between measured CGRP levels and somatic, visceral, neuropathic and inflammatory pain. These data suggest that CGRP may act as a neuromodulator in non-headache pain conditions. However, more studies are needed to fully understand the role of CGRP in nociceptive processing and therapy of chronic pain.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s10194-017-0741-2) contains supplementary material, which is available to authorized users.
Abkürzungen
CGRP
Calcitonin gene-related peptide
CGRP-LI
Calcitonin gene-related peptide – like immunoreactivity
CIP
Congenital insensitivity to pain
CNS
Central nervous system
CRPS
Complex regional pain syndrome
DRG
Dorsal root ganglia
TMJ
Temporomandibular joint

Background

The mechanism of nociception is complex involving the detection of a noxious event by nociceptors, and signal processing in the peripheral and central nervous system (CNS). Recent studies have identified specific substances and receptors with potential roles in nociception that provide therapeutic targets, including substance P, CGRP, glutamate, serotonin, TrkA receptor, vanilloid receptor and NMDA receptor [1, 2]. Chronic pain resulting from disease or injury is a major public health problem and a common complaint in general population with a lifetime prevalence ranging from 12 to 30% [3] and an enormous impact and burden on society and individuals [4]. Despite tremendous scientific effort over the past years, current pain management treatment remains suboptimal [5]. There is an unmet and urgent need for new effective therapeutic options for the management of chronic pain. Migraine manifests as pain with associated sensory disturbances and is considered as a chronic condition with episodic manifestations [6]. The role of CGRP in migraine pathophysiology has gained considerable interest in recent years [7, 8]. This led to the development of small molecule CGRP receptor antagonists for acute and preventive treatment of migraine [9, 10] and monoclonal antibodies against CGRP mechanisms for migraine prevention [11, 12].
CGRP is a 37-amino-acid neuropeptide identified in 1982 [13]. It belongs to a family of peptides including adrenomedullin, amylin and calcitonin with diverse biological functions in the periphery and in the central nervous system [14, 15]. To what extent CGRP is involved in non-headache pain conditions is not fully clarified and whether CGRP antagonism may represent a useful therapeutic approach for the treatment of chronic pain is unknown.
The aim of this systematic review was to assess the role of CGRP in non-headache pain in humans. In addition we discussed the potential role of anti-CGRP agents in the management of chronic pain.

Methods

We performed a systematic literature search identifying articles reporting original data on CGRP and non-headache pain. We concluded the literature search on Pubmed Embase and ClinicalTrials.gov on May 2016. We used the following search terms: CGRP and pain. In addition, we specified our search criteria in ClinicalTrials.gov to currently available monoclonal antibodies against CGRP (LY2951742, ALD-403, PF-04427429, LBR-101/TEV-48125) or its receptor (AMG334), and CGRP receptor antagonists (BIBN4096BS, MK-0974, MK-3207, MK-1602, MK-8825, BMS-694153, BMS-927711, BMS-742413, BI 44370 TA) [16].
Only human studies published in English language were included. Review papers editorials and other articles without original data were excluded. We also considered articles from the reference list of studies that were found to be relevant as well as literature that was known to be relevant by the authors.

Data extraction

The authors (WSS) examined the abstracts found in the literature search. Whenever the title or abstract suggested that relevant data could be part of the publication the entire article was read and discussed with the other co-authors. Studies in which patients had unclear pain history or articles without relevant data on CGRP were not included in the review.

Results

Our Pubmed Embase and ClinicalTrials search strategy identified 375 hits of which 50 studies were included in the final review (Fig. 1). After excluding 110 as duplicates, another 118 hits were excluded because these were abstracts, reviews, animal, migraine, headache studies, and incomplete studies. Subsequently, we excluded 97 studies that did not describe pain history of the patients, had no relevant data on CGRP, or had unclear methodology. In total, 50 studies were included in the final review (Fig. 1). The identified studies were further divided into five categories: 1) somatic pain, 2) visceral pain, 3) inflammatory pain, 4) neuropathic pain, and 5) clinical trials (Fig. 2).

Somatic pain

We found a total of 20 studies on the role of CGRP in somatic pain (Table 1). Using different methodological approaches and CGRP sample sources 13 studies showed higher levels of CGRP compared to controls. Eight studies directly tested for a possible correlation between pain intensity and CGRP levels. In chronic knee pain due to osteoarthrosis elevated CGRP levels were detected in serum and synovial fluid in patients compared with controls. Serum CGRP levels were positively correlated with pain intensity [17]. Chronic low back patients due to osteoarthrosis showed decreased blood CGRP levels four months after successful auricular point acupressure pain treatment compared to baseline. No decrease was found in patients who received sham treatment [18]. In addition, studies using immunofluorescence of skin biopsies reported decreased CGRP after acupuncture treatment of osteoarthrosis patients [19]. Immunohistochemistry analysis of synovial tissue from fossa acetabuli showed increased CGRP levels in patients compared to controls [20, 21]. One study reported higher levels of CGRP in hip synovium from osteoarthritis patients compared with femoral neck patients [22]. Moreover, one study [23] revealed higher levels of CGRP in synovial tissue from temporomandibular joint (TMJ) pain patients compared with controls. This study also reported positive correlation between pain and CGRP levels [23]. Biopsies from knee joint ligaments showed no difference in CGRP nerve density between patients and non-arthrosis patients [24]. In patients with osteoarthrosis CGRP concentration in cerebrospinal fluid was decreased compared to controls [25].
Table 1
Studies on the role of CGRP in somatic pain
Study
Objectives
Reported pain as part of phenotype
Method and sample size
Source of CGRP
Results
Duration of the investigated condition
Correlation between CGRP level and pain
Alpar, 2002 [1]
Determine plasma CGRP in patients with whiplash injury who were treated by carpal tunnel decompression
Chronic shoulder and neck pain due to whiplash injury
38 patients and 11 controls.
Enzyme-immunoassay kit was used to measure the plasma CGRP
Blood (plasma)
Mean plasma levels was higher in patients, 400 ng/l, than in controls, 85 ng/l. Plasma levels were reduced, 65 ng/l, after carpal tunnel decompression
NR
Reduced plasma CGRP after the operation correlated to the pain reduction
Bjur, 2005 [2]
Investigate innervation patterns of Achilles tendon in tendinosis tendon, and normal tendon
Chronic pain in tendinosis
Tissue samples from 21 patients and 9 controls
Tissue biopsies (Achilles tendon)
Inconclusive. CGRP was found in both patients and controls. The amount of CGRP-fibers was not quantified
Mean 19 months
NR
Brown, 1997 [3]
Determine density of CGRP containing sensory nerve fibers in vertebral endplate in patients with degenerative disc disease
Severe back pain with or without sciatica in degenerative disc disease
Tissue from the intervertebral discs from 15 patients undergoing anterior lumbar discectomy and 7 healthy post-mortem controls
Tissue biopsies (intervertebral discs)
Marked increase in CGRP-containing sensory nerve fibers compared with controls
NR
NR
Carlsson, 2006 [4]
Evaluate possible effects of acupuncture on sensory nerve fibers in human skin
Cervicobrachial pain, cervicocranial pain, hip pain and finger pain from arthrosis
Punch skin biopsies taken from 6 patients one week before acupuncture and 3–6 days after the 10th treatment
Tissue biopsies (skin)
The mean number of CGRP-IR nerve fibers were reduced after treatment
4 months - >10 years
NR
Danielson, 2008 [5]
Investigate CGRP prevalence in patients with tendinitis surgery.
Chronic painful patellar tendinosis
Patellar tendon biopsy in 7 patients
Tissue biopsies (patellar tendon)
CGRP rarely detected at perivascular sites
Chronic pain
NR
Dong, 2015 [6]
Examine CGRP concentrations in patients with primary knee OA and controls
Chronic knee pain from OA
Serum CGRP concentrations in OA patients (n = 65) and controls (n = 21).
Blood (serum)
CGRP levels were higher in patients, 2.43 ng/mL, than in controls, 1.95 ng/mL
NR
CGRP concentrations in serum were correlated with pain intensity
Ikeuchi, 2012 [7]
Determine sensory innervation of posterior cruciate ligament (PCL) in patients with OA
Chronic knee pain from OA
PCL samples from 10 patients and 5 pain-free controls with anterior cruciate ligament (ACL) rupture
Tissue biopsies (joint ligament)
No difference between patients and controls
NR
NR
Jonhagen, 2006 [8]
Determine CGRP in human skeletal muscle at rest and after painful eccentric exercise
Experimental muscle pain after eccentric exercise
Microdialysis catheter inserted in quadriceps muscle in 8 healthy volunteers. Samples taken before and after exercise.
Blood (plasma)
CGRP levels were higher 2 days after exercise, 5.4 fmol/ml, than directly after exercise, 4.85 fmol/ml
VAS-score was assessed on the entry day (VAS = 0), day 1 (VAS = 1) and day 3 (VAS = 2) after the exercise
CGRP concentrations was positively correlated with pain intensity (VAS)
Larsson, 1991 [9]
Investigate CGRP-levels from patients with rheumatoid arthritis and patients with meniscal/cruciate ligament injuries
Acute knee pain in meniscal/cruciate ligament injuries
Synovial fluid from the knee joint of 18 patients and 13 pain-free controls with ligament injuries
Synovial fluid (knee joint)
Increased CGRP levels in patients compared to controls.
4-27 years
NR
Lin, 2015 [10]
Investigate associations between plasma CGRP-levels and clinical outcome from APA, in patients with osteoarthritis and spinal stenosis
Chronic lower back pain (CLBP) in patients with osteoarthritis and spinal stenosis
Blood samples from 32 patients (APA-group) and 29 controls (sham APA-group). Samples were taken at baseline and 4 weeks later. VAS-score before treatment was 4.
Blood (plasma)
Patients showed a decrease in CGRP levels after treatment. No decrease in the control group
At least 3 months
56% of the patient group reported a reduction in pain, whereas only 9% controls reported a reduction
Lindh, 1999 [11]
Determine CGRP-LI levels in CSF in patients with chronic pain
Osteoarthritis, herniated lumbar disc and hip fracture pain
Sample: CSF
Subjects: 35 patients (14 had knee or hip pain, 11 had rhizopathic pain due to herniated lumbar pain, 10 had pain from hip fracture and 12 healthy controls.
Pain assessment: VAS
CSF
Decreased CGRP-LI levels were observed in patients compared to healthy controls
Osteoarthritis patients: >6 months. Rhizopathic pain: At least 1 month (1–13). Hip fracture pain: Up to 48 h
No correlation between CGRP-LI levels and VAS-values could be observed for any of the subjects participating in the study
Onuoha, 1999 [12]
Investigate CGRP levels in patients with soft tissue injury
Acute muscle and ligament pain due to injury
Plasma CGRP-concentrations in 17 patients and 15 healthy controls
Blood (Plasma)
CGRP-levels were significantly higher in patients than controls
Up to 24 h
NR
Ozawa, 2006 [13]
To determine sensory fibers innervating human degenerated lumbar intervertebral discs
Discogenic low back pain
Lumbar intervertebral disc was harvested from 8 patients, and immunostained for CGRP
Tissue biopsies (intervertebral disc)
CGRP-IR nerve fibers were observed in 6 out of 8 patients
NR
NR
Samuelsson, 1993 [14]
Determine CSF CGRP levels in cancer patients
Cancer pain
CSF from 10 patients compared with 10 controls
CSF
No difference in CGRP-levels between patients and controls
NR
No difference between patients with pain and controls
Sasaki, 2013 [15]
Investigate innervation patterns of ECRB in patients with recalcitrant tennis elbow
Lateral epicondylitis
Tissue biopsies from 8 patients and 2 controls. The control group suffered from osteochondritis
Tissue biopsies (lateral epicondyle)
A decrease in the immunorectivity of CGRP compared to controls
Mean duration 23 months
The innervation pattern did not appear to be correlated with VAS-score
Sato, 2004 [16]
Elucidate expression of CGRP in temporomandibular joint (TMJ) from patients with internal derangement
TMJ pain
Synovial fluid from 48 patients and 7 controls, who had pain-free habitual dislocation
Synovial fluid (TMJ)
Increased CGRP in patients compared to controls
Mean duration 6 months
Positive correlation between the extent score of CGRP-levels and joint pain
Saxler, 2007 [17]
Determine presence of CGRP-immunopositive nerve fibers in patients with OA
Hip pain from OA
Soft tissue biopsies from fossa acetabuli in 3 patients and
6 pain-free controls. 3 controls had a failed THA and 3 controls had femoral neck fractures
Tissue biopsies (fossa acetabuli)
Increased CGRP-LI in patients compared to controls.
NR
Positive correlation between CGRP and pain
Takeshita, 2012 [18]
Clarify sensory innervation and inflammatory cytokines in OA patients
Severe hip pain from OA
Synovium from 50 patients and 12 controls with femoral neck fracture
Synovial fluid (hip)
CGRP-IR sensory nerve fibers were observed in 54% of the patients and 0% in controls
NR
NR
Takeuchi, 2007 [19]
Determine CGRP’s role in patients with lumbar disc herniation, before and after lumbar discectomy
Sciatic pain/lumbar disc herniation
Plasma CGRP was measured in 27 patient before and 3 weeks after lumbar discectomy
Blood (plasma)
Plasma CGRP-levels were reduced after lumbar discectomy
3 weeks
Reduced plasma CGRP after the operation correlated to lower VAS-levels
Wang, 2015 [20]
Explore mechanisms of possible involvement and regulation of CGRP in pathological and inflammatory processes of arthritis in patients with developmental dysplasia of the hip (DDH)
Hip pain from OA
Synovial tissue samples from 67 patients: 35 with moderate DDH and 32 patients with severe DDH.
15 controls with traumatic femoral fracture
Synovial tissue (fossa acetabuli)
Increased CGRP in synovium fluid from patients in the severe DDH group compared to the moderate DDH group and controls
NR
The highest amount of CGRP correlated with the highest VAS
APA Auricular point acupressure, CGRP Calcitonin gene-related peptide, CGRP-LI Calcitonin gene-related peptide-like immunoreactivity, CLBP Chronic low back pain, CRPS Complex regional pain syndrome, CSF Cerebrospinal fluid, ECRB Extensor carpi radialis brevis, KL grades Kellgreen and Lawrence classification, used to assess the severity of OA, NR not reported, OA Osteoarthritis, PHN Postherpetic neuralgia, THA Total hip arthroplasties
In patients suffering from chronic pain due to degenerative disc disease disc biopsies showed increased CGRP compared to post-mortem control discs [26]. Biopsies from intervertebral discs in patients with low back pain contained CGRP-IR nerve fibers [27].
Patients suffering from shoulder and neck pain due to whiplash injury were found to have higher blood CGRP levels compared to controls [28]. Another study of patients with disc herniation pain reported increased blood levels of CGRP which were normalized after discectomy [29]. Blood CGRP levels were also elevated in patients with soft tissue injury (i.e. muscle or ligament pain) compared with controls [30]. Furthermore, one radioimmunoassay study of knee synovial fluid from patients with meniscal or ligament injury revealed higher CGRP levels compared to controls [31].
Immunohistochemistry analysis of biopsies of Achilles tendons from patients with chronic painful tendinosis showed no changes in CGRP levels in patients compared to controls [32] while another study in patients with patellar tendinosis found the presence of CGRP, but had no control group [33]. One study reported decreased CGRP in the extensor carpi radialis brevis tendon biopsy from patients with tennis elbow compared to patients with osteochondritis [34].
Samuelsson and colleagues [35] compared CGRP levels in cerebrospinal fluid from cancer patients with pain and found no difference between patients and non-pain control patients. A microdialysis study in the vastus lateralis of the quadriceps muscles before and during pain after eccentric exercise (repetitive muscle contractions while the muscle is lengthening under load) reported increased CGRP levels during pain compared with baseline [36].

Visceral pain

Eight studies examined CGRP in different types of visceral pain conditions (Table 2).
Table 2
Studies on the role of CGRP in visceral pain
Study
Objectives
Reported pain as part of phenotype
Method and sample size
Source of CGRP
Results
Duration of the investigated condition
Correlation between CGRP level and pain
Arellano, 2011[1]
Investigate nerve growth factor role in development of pelvic pain in patients with endometriosis
Pain from endometriosis
Peritoneal fluids from 65 patients, 54 with pain, 11 without pain.
22 controls, where 12 reported pelvic pain
Peritoneal fluid
CGRP-neurite outgrowth was seen in patients
NR
The CGRP-neurite outgrowth did not correlate with pain symptoms
Büchler, 1992 [2]
Identify characteristics of peptidergic innervation in patients with chronic pancreatitis
Pain from chronic pancreatitis
Pancreatic tissue from 20 patients compared to 10 organ donors
Tissue biopsies
(pancreatic tissue)
CGRP-immunostaining was intensified in patients
NR
NR
Mönnikes, 2005 [3]
Assess whether functional dyspepsia (FD) patients have altered mucosal CGRP concentrations
Pain from functional dyspepsia
Gastric mucosal biopsies from 13 patients and 18 controls.
Biopsies were taken during gastric distention
Tissue biopsies
(gastric mucosa)
No difference in CGRP-levels between patients and controls
The gastric distention took up to 80 min
A negative correlation between CGRP concentrations and pain was observed in patients.
No such correlation was found in controls
Tokushige, 2006 [4]
Determine the nerve fibers in patients with peritoneal endometriosis
Pain from endometriosis
Peritoneal endometriotic tissue from 40 patients and 36 healthy controls. Also 9 specimens from endosalpingiosis lesions were prepared
Tissue biopsies
(endometriotic tissue)
Increase of CGRP-nerve fibers in patients, compared to controls and endosalpingiosis lesions
NR
NR
Tokushige, 2007 [5]
Investigate types of nerve fibers in endometrium and myometrium in women with endometriosis
Pain from endometriosis
Tissue biopsies from 10 patients and 35 controls.
All tissue biopsies were taken during hysterectomy
Tissue biopsies
(endometriotic tissue)
Increased nerve fiber densities compared to controls
NR
NR
Tympanidis, 2003 [6]
Evaluate nerve fiber density and pattern in patients with vulvodynia
Pain from vulvodynia
Biopsies from the wall of the vulval vestibule from 12 patients and 8 controls
Tissue biopsies
(vulval vestibule)
No difference in CGRP-immunostaining between patients and controls
NR
NR
Währborg, 1999 [7]
Clarify potential involvement of CGRP in anginal pain and myocardial ischemia in humans
Chest pain from angina and acute myocardial infarction
Plasma from 87 patients with AMI compared to 14 patients with severe angina pectoris
Blood
(plasma)
No difference in CGRP-levels between patients with AMI and angina pectoris
At least 15 min
No correlation between CGRP-levels and pain
Yoshida, 2013 [8]
Estimate expression of CGRP in esophageal mucosa in nonerosive reflux disease (NERD) patients
Pain due to NERD
Biopsies from 24 patients, compared to 24 controls
Tissue biopsies
(esophageal mucosa)
No difference in CGRP-levels between patients and controls
NR
NR
Immunofluorescence-based analysis of peritoneal fluid obtained during diagnostic laparoscopy in patients with endometriosis showed increased CGRP levels compared to peritoneal fluid from controls without endometriosis [37]. Visual analogue scale scores were registered in all patients but authors found no correlation between CGRP levels and severity of pain. Immunohistochemistry analyses of peritoneal endometriotic lesions and normal peritoneum from non-endometriotic women showed increased CGRP in affected tissue material [38]. Using the same technique, increased CGRP levels were found in endometrium and myometrium in women with, but not in those without endometriosis. Pain measurement data was not reported [39]. CGRP levels were also studied in patients with vulvodynia. Analysis of vulval vestibule tissue revealed no differences in CGRP levels between patients with vulvodynia and controls [40].
Gastric mucosal biopsies from patients with non-erosive reflux disease [41] and functional dyspepsia [42] were investigated with enzyme- and radioimmunoassay. None of the studies found differences in CGRP levels between patients and controls but a negative correlation between CGRP concentrations and pain scores was reported in the latter [42]. CGRP has also been investigated with immunohistochemistry in patients with alcohol-based painful chronic pancreatitis and increased CGRP levels in patients were reported compared with pancreatic tissue from organ donors [43].
Plasma CGRP levels were studied in patients with suspected or definite acute myocardial infarction at admission at a coronary care unit [44]. This study revealed no difference in CGRP levels between patients with and without acute myocardial infarction and no difference between patients with pain and those without pain.

Inflammatory pain

Eight studies on CGRP and inflammatory pain conditions were identified (Table 3). ELISA of dermal microdialysate from volar forearm showed elevated blood CGRP levels in ten healthy volunteers with capsaicin-induced pain [45]. No CGRP release was detected via dermal microdialysate after electrical stimulation in the same area. Correlation between pain intensity or threshold and CGRP concentration was not tested [45]. In contrast another study found CGRP in the dialysate after histamine iontophoresis, but not after capsaicin application in the volar forearm [46]. One study performed immunohistochemistry of skin biopsies after intradermal capsaicin injection and reported complete loss of CGRP visualization 72 h after injection [47].
Table 3
Studies on the role of CGRP in inflammatory pain
Study
Objectives
Reported pain as part of phenotype
Method and sample size
Source of CGRP
Results
Duration of the investigated condition
Correlation between CGRP level and pain
Geber, 2007 [1]
Evaluate pain, hyperalgesia and neurosecretory function in pain models with CAP and ES
Experimental pain: CAP and ES
Samples from dermal microdialysis taken from 10 healthy volunteers. Patients rated pain levels after CAP and ES stimulation
Blood
(plasma)
CGRP increase was measured after CAP, not after ES
2 h
NR
Hamed, 2011 [2]
Determine cutaneous innervation in burn patients with chronic pain
Chronic inflammatory skin pain
Skin biopsies from 12 patients and 33 controls suffering from unilateral injury, without pain
Tissue biopsies
(skin)
Increase in CGRP density compared to controls
>24 months
CGRP-levels were higher in patients with pain compared to controls
Krämer, 2005 [3]
Explore effect of specific blockers of NEP (phosphoramidon) and ACE (captopril) on intensity of neurogenic inflammation
Experimental pain: ES
Samples from dermal microdialysis were taken from 8 healthy volunteers.
Patients quantified pain sensation during electrical stimulation using VAS
Dermal microdialysis
CGRP release could be measured after phosphoramidon perfusion
1 h
CGRP release did not correlate to pain ratings during phosphoramidon infusion
Kwak, 2014 [4]
Evaluate CGRP’s effect on wound healing process in hypertrophic scar formation
Inflammatory pain in scars
Skin biopsies from 43 patients. Biopsies were taken from scars, and also from a normal skin area
Tissue biopsies
(skin)
Increased CGRP-levels in scars compared to matched unburned skin
>12 months
Increased CGRP-levels in painful scar areas compared to normal skin
Onuoha, 2001 [5]
Examine plasma CGRP levels in patients with burns
Inflammatory pain from burn
Plasma was obtained from 13 patients immediately on hospital admission and 24 h after admission. 13 volunteers served as controls
Blood
(plasma)
CGRP levels were higher on admission, 4.9 pmol/L and after 24 h, 7.3 pmol/L, than in controls, 1.9 pmol/L
NR
NR
Salomon, 2008 [6]
Evaluate CGRP-levels in AD patients during exacerbation and disease remission
Pruritus due to AD
Plasma from 49 patients and 32 healthy controls
Blood
(plasma)
CGRP-levels were lower compared to healthy controls
Mean 20.75 years (1-55years)
High CGRP concentrations correlated with
severe pruritus
Schmelz, 1997 [7]
Examine neuropeptide release in human skin elicited by histamine iontopheresis and topical CAP application
Experimental pain: histamine iontopheresis and CAP application
Samples from dermal microdialysis taken from 10 healthy volunteers.
Patients were pain free prior start
Dermal microdialysis
CGRP concentration increased after histamine iontophoresis, but not capsaicin application
3 h
NR
Simone, 1998 [8]
Determine whether hyperalgesia after intradermal injection of CAP could be attributed to morphological changes in ENF’s
Experimental pain: intradermal CAP injection
Skin biopsies from 8 healthy volunteers
Tissue biopsies
(skin)
Complete loss of CGRP-fibers was observed 72 h after capsaicin injections. They reappeared 3–4 weeks after
6 weeks
NR
ACE Angiotensin-converting enzyme, AD Atopic dermatitis, CAP Capsaicin injection, ENF’s Epidermal nerve fibers, ES Electrical current stimulation, NEP Neutral endopeptidase
Using the ELISA and dermal microdialysis method in healthy volunteers CGRP release was reported after electrical stimulation upon phosphoramidon but not after captopril infusion in the volar forearm [48]. Phosphoramidon and captopril, respectively, inhibit neutral endopeptidase and angiotensin-converting hormone, which are both involved in neuropeptide degradation [49].
Immunohistochemical analysis of skin biopsies in patients with painful scars from burn showed increased CGRP compared with controls with burn scars without pain [50]. Another study reported increased CGRP in hypertrophic burn scar compared to biopsies from unburned scars. Pain intensity was higher in patients with burn scars [51]. Moreover ELISA of peripheral blood showed increased CGRP levels up to 24 h after burn injuries compared with healthy volunteers [52] and in patients with pruritus due to atopic dermatitis [53]. Furthermore, CGRP levels were positively correlated with the severity of pruritus [53]. Nociceptive fibers have been shown to be involved in the sensation of pruritus [54].

Neuropathic pain

We identified 11 studies in this category (Table 4). Radioimmunoassay showed higher serum CGRP levels in 19 patients with complex regional pain syndrome (CRPS) compared to controls. The difference was normalized after a 9-month pain management therapy [55]. In contrast another study found decreased serum CGRP levels in chronic CRPS patients (n = 12) compared with healthy controls [56]. No correlation between pain and CGRP levels was found in either study [55, 56]. Moreover, immunofluorescence analysis of skin biopsies from amputated limbs in CRPS patients showed loss of CGRP expression in two patients compared with skin biopsies from five controls. Correlation between pain measures and CGRP levels was not tested [57]. In post-herpetic neuralgia, increased CGRP expression in the affected skin compared with skin from a contralateral side in the same patient was reported by using immunofluorescence analyses of skin biopsies [58]. In one study using immunofluorescence of skin biopsies [59] no difference in CGRP expression was found between patients with chronic pain due to nerve injury after hand surgery and controls. The immunohistochemistry of peripheral nerve biopsies harvested from patients with Morton’s neuroma, which results in neuropathic pain, showed increased amount of CGRP in patients compared with controls [60].
Table 4
Studies on the role of CGRP in neuropathic pain
Study
Objectives
Reported pain as part of phenotype
Method and sample size
Source of CGRP
Results
Duration of the investigated condition
Correlation between CGRP level and pain
Albrecht, 2006 [1]
Investigate CGRP expression in skin from amputated extremity affected by CRPS
CRPS after amputation in upper and lower limbs
Skin tissue from 2 patients and 5 controls
Tissue biopsies
(skin)
Loss of CGRP expression in CRPS patients
NR
NR
Attal, 2016 [2]
Determine CGRP levels in peripheral neuropathic pain patients after treatment with botulinum toxin A
Peripheral neuropathic pain, mixed group
ELISA of biopsy from 23 patients with active treatment and 17 patients with placebo treatment at week 1 and 4 after study start
Skin
No change in CGRP levels at week 4 compared to week 1. Average pain score was not changed either
NR
None
Awawdeh, 2002 [3]
Investigate presence of CGRP in the gingival crevicular fluid of teeth diagnosed with pain of pulpal origin
Tooth pain
Gingival crevicular fluid from a painful and non-painful site from 54 patients undergoing pulpectomy. 1 week after fluid was collected from 21 patients
Gingival crevicular fluid
No difference in CGRP-levels between painful tooth compared to the contralateral control tooth
NR
No clear association between CGRP-levels and dental pain
Boras, 2010 [4]
Determine saliva and serum CGRP levels in patients with BMS
Burning mouth syndrome
Saliva and serum from 26 patients and 22 controls
Saliva and serum
No difference between patients and controls
NR
NR
Birklein, 2001 [5]
Test contribution of neuropeptide release to pathophysiology of CRPS
CRPS in upper or lower limbs
Serum CGRP concentrations were measured in 19 patients on the affected (n = 19) side and non-affected side (n = 13) before and 9 months after therapy (n = 9). Comparison with controls (n = 16)
Blood
(serum)
Increased CGRP levels in CRPS patients.
No difference in CGRP levels in blood taken from affected versus non-affected side.
After therapy – normalization of CGRP levels compared to healthy controls
Mean 29 weeks (range: 2 to 188 weeks)
No correlation between CGRP-levels and pain
Chavarría-Bolanos, 2014 [6]
Determine CGRP-levels in dental pulp tissue samples from 8 patients subjected to undergo controlled orthodontic intrusive forces
Tooth pain
Human premolar dental pulp tissue was extracted from 8 patients, and 8 controls
Tissue biopsies
(dental pulp)
No differences in CGRP levels between the two groups
24 h
NR
Hou, 2011 [7]
Determine whether CGRP-IL is increased among epidermal keratinocytes in PHN and diabetes
PHN and small fiber neuropathy
Punch biopsies from 5 patients with PHN from painful areas, 5 patients with diabetes (biopsies from feet) and 11 controls
Tissue biopsies
(skin)
Increased CGRP-levels in keratinocytes from PHN patients compared to controls
NR
CGRP levels were higher in painful skin areas compared to non-painful locations
Kalliomäki, 2011 [8]
Investigate structural and functional differences between patients with and without chronic pain following nerve injury
Hand pain due to nerve injury
Skin biopsies from 21 patients with pain and 9 controls without pain. All participants required hand surgery
Tissue biopsies
(skin)
No difference in CGRP-staining between patients and controls
>1 year
No significant difference between pain and non-pain patients
Lindqvist. 2000 [9]
Examine CGRP expression in painful Morton’s neuroma patients
Forefoot Morton’s neuroma
11 nerve biopsies from 8 patients and 4 controls
Nerve biopsies
Increased levels of CGRP-IR nerve fibers compared to controls
NR
NR
Schinkel, 2009 [10]
Compare systemic inflammatory mediators in patients with CRPS I with controls
CRPS in the upper limb
Blood samples were obtained from 22 patients. 12 patients had chronic CRPS and 10 patients had acute CRPS. Patients were compared to 8 controls
Blood
(plasma)
CGRP-levels were lower in chronic CRPS patients, compared to controls
Acute: < 6 months
Chronic: > 6 months
NR
Zidverc-Trajkovic, 2009 [11]
Determine saliva CGRP levels in patients with BMS
Burning mouth syndrome
Saliva from 78 patients and 16 healthy controls
Saliva
Inconclusive, CGRP levels were non-significantly decreased in comparison to controls
At least 6 months
NR
BMS Burning mouth syndrome, CRPS Complex regional pain syndrome, CSF Cerebrospinal fluid
Four studies reported on CGRP radioimmunoassay: 1) gingival crevicular fluid in unilateral tooth pain patients [61] 2) saliva from burning mouth syndrome patients [62, 63], and 3) pulp biopsy in patients undergoing orthodontic intrusion [64]. None of the studies reported alteration in CGRP expressions in painful sides when compared with the non-painful side [61] or with controls [62, 64].
Attal et al. [65] investigated 152 patients with peripheral neuropathic pain of whom 68 were treated with botulinum toxin A and 66 received placebo. CGRP was analyzed in skin biopsies using ELISA at week 1 and 4 in 23 patients who received botulinum toxin A and in 17 patients who received placebo. No difference between groups was found [65].

CGRP antagonists and antibodies and clinical trials

We did not identify any clinical trials on CGRP antagonists and antibodies for the treatment of non-headache pain by searching PubMed and Embase. Search on ClinicalTrial.gov for current CGRP antagonists and antibodies for the treatment of non-headache pain only yielded three studies.
The acute effect of PF-04427429 anti-CGRP monoclonal antibody, on attenuation of flare response after capsaicin challenge, used to induce experimental human pain, was studied in a double blind, randomized, placebo-controlled, third-party open, modified cross-over study in male healthy volunteers and using EMLA® cream as positive control [66]. However, primary outcome measure of the study was mean blood perfusion induced by capsaicin challenge (results not reported on ClinicalTrials.gov) and no pain perception measures were studied.
A phase 2 randomized, double-blind, placebo and active-controlled trial in patients with mild to moderate osteoarthritis knee pain failed to demonstrate efficacy of LY2951742, monoclonal antibody to CGRP [67]. The study was terminated. A total of 266 patients were randomized to 1 of 6 treatment arms: LY2951742 5 mg, 50 mg, 120 mg, or 300 mg, celecoxib 200 mg, or placebo. Using a Bayesian dose–response longitudinal model, response rates to all four LY2951742 treatment arms were not different from placebo while celecoxib met criteria for a positive study [68].
An ongoing study on remote ischemic conditioning in patients with ulcerative colitis a condition associated with abdominal pain and diarrhea is still in a recruiting phase [69]. Investigators plan to study changes of serum and mucosal CGRP levels (secondary endpoints) in patients with ischemic colitis after remote ischemic conditioning, a repeated brief and non-harmful suppression of blood circulation induced by placing a blood pressure cuff around the right or left arm.

Discussion

Summary of findings

The present review revealed the association between measured CGRP levels and somatic visceral, neuropathic and inflammatory pain. We found that in somatic pain conditions in particular, CGRP levels correlated with pain. Increased CGRP levels were reported in plasma, synovial and cerebrospinal fluid, tissue biopsies in individuals with degenerative disc disease, osteoarthritis and TMJ-pain. Furthermore, CGRP was elevated in acute pain conditions and pain after exercise.
In total 13 out of 20 studies on somatic pain increased levels of CGRP were reported. Five studies showed no difference or had no control group. Four out of eight studies investigated CGRP in experimental models of inflammatory pain. The remaining four studies reported elevated CGRP levels in patients with pain caused by scars and pruritus. There was no consensus regarding correlation between neuropathic pain and CGRP levels. Six out of eleven studies showed no difference in CGRP levels, three studies reported a positive correlation, and two studies reported a negative correlation between neuropathic pain and CGRP levels. In visceral pain conditions a correlation between gynecological pain and high CGRP levels were found in tissue biopsies and peritoneal fluid. However, only two studies used a control group or control conditions.
Thirty out of fifty studies (60%) included controls and suggested an association between CGRP levels and the respective pain condition. Twenty-six (52%) studies reported a positive association whereas four studies (8%) reported decreased CGRP levels. Studies reporting positive association investigated blood (10 studies), skin (5 studies), synovial tissue/fluid (5 studies), and other affected tissues (6 studies). Collectively, these studies showed a positive correlation between high CGRP levels and somatic pain conditions, especially osteoarthritis, acute muscular pain and chronic joint/muscular pain. These findings raise two important questions: what is the role of CGRP in the transmission of nociceptive signals and whether CGRP causes or modulates pain?

CGRP and pathophysiology of pain

CGRP is widely distributed in the peripheral and central nervous system [70, 71] and CGRP receptors are expressed in pain pathways [7276]. CGRP-like immunoreactivity (CGRP-LI) is found in 40–50% of dorsal root ganglia (DRG) neurons [77]. CGRP-LI was found C-fiber (46%), delta-fiber (33%), and A-alpha/beta fiber (17%) neurons [77]. Moreover, CGRP is usually co-localized with other neuropeptides, including substance P [78] and neurokinins [79] in DRG neurons. Peripheral CGRP-LI fibers terminate in lamina I, III and V of spinal cord [80] and CGRP-containing DRG neurons innervate joints [81]. Thus, CGRP and its receptors are widely distributed in peripheral and central pain pathways.
In animals CGRP can be released from peripheral and central nerve endings upon noxious pain mechanical stimulation of the skin [8285]. In rats, the major part of circulating CGRP is released from perivascular nerve terminals [86, 87]. Acute and chronic nociception leads to altered release of CGRP from sensory nerve endings and central terminals into the dorsal horn of the spinal cord [8891]. In rats, CGRP applied spinally causes facilitation of central excitability and central sensitization [92, 93]. Kessler et al. [94] demonstrated reduced mechanical allodynia in an animal model of OA following administration of an intrathecal CGRP receptor antagonist [94]. Animal in vitro studies reported direct activation of nociceptors by CGRP [95, 96]. CGRP injected into mouse hind paw skin produced mechanical allodynia [97]. In humans, however, a direct activation of nociceptive fibers is unlikely. CGRP injected intradermally or intramuscularly did not produce pain [98].
CGRP is also found in free nerve endings in skin and synovium) and perivascular afferents in different structures in both humans and animals [99101]. The release of CGRP from these fibers causes vasodilation suggesting a role in neurogenic inflammation [98, 101, 102]. The question is whether CGRP exerts either pro- or anti-inflammatory/nociceptive effects. It is possible that CGRP release reflects the response of the nocifensor system to injury and inflammation to evoke protective vasodilatation. Deficiency of alpha CGRP (αCGRP knockout mice) was associated with enhanced inflammatory responses in the hippocampus and hypothalamus and reduced the survival rate compared to wild-type mice in septic shock condition [103]. However, αCGRP knockout mice displayed lower pain sensitivity to heat stimulation faster accumulation of c-Fos compared to wild-type animals after incision and complete Freund’s adjuvant injection [104]. In animals, sustained CGRP release may induce peripheral sensitization [105] likely due to release of inflammatory mediators (bradykinin, prostaglandins, etc.) from nerve endings and cells of immune system [106108].
Inflammatory diseases of the joints tendons and discs may be associated with elevated levels of CGRP (Additional files 1 and 2: Tables S1 and S2). These data suggest that abnormal release of CGRP could be a marker of sensory afferent activation. Comparing CGRP changes in different tissue materials (i.e. blood, synovium, skin, CSF, ligament tissue, mucosa, etc.), it seems that elevated CGRP is more frequently found in blood, synovium and skin. Bullock et al. [109] suggested that CGRP release during joint degeneration in osteoarthritis might play an important role in the peripheral sensitization and proposed possible analgesic effect of CGRP antagonists in this condition. CGRP stimulates proliferation and migration of human endothelial cells [110], causing angiogenesis with the co-localized CGRP-containing perivascular nerve fibers. Intra-articular growth of CGRP-containing perivascular nociceptors have been reported in patients with osteoarthritis. It has further been shown that nociceptive nerve fibers innervating joints are sensitized in these patients [111] contributing to the experience of pain. Immunohistochemistry of forearm skin biopsies in patients with congenital insensitivity to pain (CIP) showed reduced amount of CGRP compared to controls [112]. Thus, measurement of CGRP may be regarded a marker of sensory afferent activation in the respective tissue during a pain condition [113]. This indicates that CGRP not only contributes to proliferation of CGRP-containing nociceptors, but could sensitize these nociceptors via neurogenic inflammation in humans. Whether CGRP causes pain per se can be examined by application of exogenous CGRP. Interestingly, dose-dependent angiogenesis after intra-articular CGRP injection in the rat knee can be blocked by the CGRP receptor antagonist, BIBN4096BS [114]. One way of exploring this hypothesis would be to study CGRP levels in humans after exposure to painful stimuli. In healthy volunteers, intradermal capsaicin injections produced a steady increase of CGRP levels in the first sampling period, but failed to reach significance in the second session [45]. The latter could be explained by capsaicin-induced desensitization of neuropeptide release from primary afferents [115]. Another study demonstrated that capsaicin-induced vasodilation in the human skin was mainly mediated by CGRP and not by other substances with vasodilator properties including prostaglandins, nitric oxide, or substance P [116]. Only few studies have investigated the effect of CGRP antagonist after intradermal capsaicin injections [66, 117]. Chi-Chung Li et al. [117] reported that CGRP antagonist MK-3207 inhibited capsaicin-induced vasodilation in skin. Sinclair et al. [118] demonstrated reduced increase in dermal blood flow after topical capsaicin application in the forearm of healthy volunteers who were pretreated with CGRP antagonist (telcagepant). The degree of inhibition in capsaicin-induced dermal blood flow was shown to be increased with higher LY2951742, CGRP monoclonal antibody, plasma concentrations suggesting dose–response relationship [119].
While increased CGRP levels in the affected tissue and synovial material indicate ingrowth of pain sensitive nerve fibers in the tissue it is unclear why CGRP level increases in blood and skin. CGRP is synthesized in central and peripheral neurons [120]. Two studies investigated CGRP levels in the cerebrospinal fluid during pain and found 1) no difference in cancer pain patients compared to controls [35], and 2) low CGRP levels in osteoarthrosis patients [25]. In contrast, biochemical studies in osteoarthrosis patients reported a positive association between pain and CGRP levels in blood [17, 18], synovial material [2022], and skin [19]. Dermal electrical current stimulation in humans caused increased CGRP in blood [48]. However, a recent study randomized, double-blind, placebo and active-controlled study in patients with osteoarthritis knee pain did not demonstrate efficacy of LY2951742, monoclonal antibody to CGRP against placebo and the trial was terminated [68]. However, the study was only done in patients with mild and moderate symptoms. It is possible that patients with severe osteoarthritis involving other joints may respond differently. Other factors that may confound the results include the long duration of the disease (not reported in abstract), which can indicate presence of central sensitization and level of activity of patients that may worsen symptoms including pain. No studies to date have investigated the efficacy of monoclonal antibodies against CGRP receptor in patients with osteoarthritis knee pain.
Further studies addressing these issues are warranted.

Conclusions

The present review suggests that CGRP may play a role in pain transmission in somatic pain conditions such as joint and muscular chronic pain. CGRP might have a pro-inflammatory role in peripheral nervous system by leading to release of pro-nociceptive substances and by facilitating central nociceptive transmission and contributing to central sensitization. However, the exact mechanisms and involvement of CGRP in nociceptive processing are not fully clarified. Understanding these mechanisms may lead to the potential development of new pharmacotherapies targeting CGRP and its receptors. Efficacy and safety of the CGRP antagonists and antibodies has already been established in migraine and this paves the way for more clinical trials in non-headache pain conditions.

Funding

We thank the Lundbeck Foundation (R155–2014–171).

Authors’ contributions

WSS conducted the literature search. All authors contributed with data interpretation, drafting and revision of the manuscript. All authors read and approved the final manuscript.

Competing interests

WSS, SA, and FMA report no conflicts of interest in relation to this paper. PJG reports personal fees from Amgen, during the conduct of the study; PJG also reports grants and personal fees from Allergan and eNeura, and personal fees from Autonomic Technologies, Bristol-Myers Squibb, Alder Biopharmaceuticals, Pfizer, Impax, Dr Reddy’s Laboratories, Zosano, CoLucid, Eli Lilly, Medtronic, Avanir, Gore, Heptares, NuPathe, and Teva Pharmaceuticals. MA reports personal fees from Alder Biopharmaceuticals, Allergan, Amgen, Autonomic Technologies (ATI), Eli Lilly and Teva Pharmaceuticals, outside the submitted work; MA is also a principal investigator (PI) for Amgen trials 20120178, 20120295, 20130255, and 20120297, and GM-11 gammaCore-R trial.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
1.
3.
Zurück zum Zitat Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D (2006) Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain 10:287–333PubMedCrossRef Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D (2006) Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain 10:287–333PubMedCrossRef
4.
Zurück zum Zitat McNamee P, Mendolia S (2014) The effect of chronic pain on life satisfaction: evidence from Australian data. Soc Sci Med 121:65–73PubMedCrossRef McNamee P, Mendolia S (2014) The effect of chronic pain on life satisfaction: evidence from Australian data. Soc Sci Med 121:65–73PubMedCrossRef
5.
Zurück zum Zitat Takai Y, Yamamoto-Mitani N, Abe Y, Suzuki M (2015) Literature review of pain management for people with chronic pain. Jpn J Nurs Sci 12:167–183PubMedCrossRef Takai Y, Yamamoto-Mitani N, Abe Y, Suzuki M (2015) Literature review of pain management for people with chronic pain. Jpn J Nurs Sci 12:167–183PubMedCrossRef
6.
8.
Zurück zum Zitat Ho TW, Edvinsson L, Goadsby PJ (2010) CGRP and its receptors provide new insights into migraine pathophysiology. Nat Rev Neurol 6:573–582PubMedCrossRef Ho TW, Edvinsson L, Goadsby PJ (2010) CGRP and its receptors provide new insights into migraine pathophysiology. Nat Rev Neurol 6:573–582PubMedCrossRef
9.
Zurück zum Zitat Lassen LH, Haderslev PA, Jacobsen VB, Iversen HK, Sperling B (2002) CGRP may play a causative role in migraine. Cephalalgia 22:54–61PubMedCrossRef Lassen LH, Haderslev PA, Jacobsen VB, Iversen HK, Sperling B (2002) CGRP may play a causative role in migraine. Cephalalgia 22:54–61PubMedCrossRef
10.
Zurück zum Zitat Sun H, Dodick DW, Silberstein S, Goadsby PJ, Reuter U, Ashina M, Saper J, Cady R, Chon Y, Dietrich J, Lenz R (2016) Safety and efficacy of AMG 334 for prevention of episodic migraine: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol 15:382–390PubMedCrossRef Sun H, Dodick DW, Silberstein S, Goadsby PJ, Reuter U, Ashina M, Saper J, Cady R, Chon Y, Dietrich J, Lenz R (2016) Safety and efficacy of AMG 334 for prevention of episodic migraine: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol 15:382–390PubMedCrossRef
11.
Zurück zum Zitat Bigal ME, Dodick DW, Rapoport AM, Silberstein SD, Ma Y, Yang R, Loupe PS, Burstein R, Newman LC, Lipton RB (2015) Safety, tolerability, and efficacy of TEV-48125 for preventive treatment of high-frequency episodic migraine: A multicentre, randomised, double-blind, placebo-controlled, phase 2b study. Lancet Neurol 14:1081–1090PubMedCrossRef Bigal ME, Dodick DW, Rapoport AM, Silberstein SD, Ma Y, Yang R, Loupe PS, Burstein R, Newman LC, Lipton RB (2015) Safety, tolerability, and efficacy of TEV-48125 for preventive treatment of high-frequency episodic migraine: A multicentre, randomised, double-blind, placebo-controlled, phase 2b study. Lancet Neurol 14:1081–1090PubMedCrossRef
12.
Zurück zum Zitat Dodick DW, Goadsby PJ, Silberstein SD, Lipton RB, Olesen J, Ashina M, Wilks K, Kudrow D, Kroll R, Kohrman B, Bargar R, Hirman J, Smith J, ALD403 study investigators (2014) Safety and efficacy of ALD403, an antibody to calcitonin gene-related peptide, for the prevention of frequent episodic migraine: A randomised, double-blind, placebo-controlled, exploratory phase 2 trial. Lancet Neurol 13:1100–1107CrossRef Dodick DW, Goadsby PJ, Silberstein SD, Lipton RB, Olesen J, Ashina M, Wilks K, Kudrow D, Kroll R, Kohrman B, Bargar R, Hirman J, Smith J, ALD403 study investigators (2014) Safety and efficacy of ALD403, an antibody to calcitonin gene-related peptide, for the prevention of frequent episodic migraine: A randomised, double-blind, placebo-controlled, exploratory phase 2 trial. Lancet Neurol 13:1100–1107CrossRef
13.
Zurück zum Zitat Amara SG, Jonas V, Rosenfeld MG, Ong ES, Evans RM (1982) Alternative RNA processing in calcitonin gene expression generates mRNAs encoding different polypeptide products. Nature 298:240–244PubMedCrossRef Amara SG, Jonas V, Rosenfeld MG, Ong ES, Evans RM (1982) Alternative RNA processing in calcitonin gene expression generates mRNAs encoding different polypeptide products. Nature 298:240–244PubMedCrossRef
14.
Zurück zum Zitat Brain SD, Cambridge H (1996) Calcitonin gene-related peptide: vasoactive effects and potential therapeutic role. Gen Pharmacol 27:607–611PubMedCrossRef Brain SD, Cambridge H (1996) Calcitonin gene-related peptide: vasoactive effects and potential therapeutic role. Gen Pharmacol 27:607–611PubMedCrossRef
15.
Zurück zum Zitat Sexton PM (1991) Central nervous system binding sites for calcitonin and calcitonin gene-related peptide. Mol Neurobiol 5:251–273PubMedCrossRef Sexton PM (1991) Central nervous system binding sites for calcitonin and calcitonin gene-related peptide. Mol Neurobiol 5:251–273PubMedCrossRef
16.
Zurück zum Zitat Vécsei L, Szok D, Csáti A, Tajti J (2015) CGRP antagonists and antibodies for the treatment of migraine. Expert Opin Investig Drugs 24:31–41PubMedCrossRef Vécsei L, Szok D, Csáti A, Tajti J (2015) CGRP antagonists and antibodies for the treatment of migraine. Expert Opin Investig Drugs 24:31–41PubMedCrossRef
17.
Zurück zum Zitat Dong T, Chang H, Zhang F, Chen W, Zhu Y, Wu T, Zhang Y (2015) Calcitonin gene-related peptide can be selected as a predictive biomarker on progression and prognosis of knee osteoarthritis. Int Orthop 39:1237–1243PubMedCrossRef Dong T, Chang H, Zhang F, Chen W, Zhu Y, Wu T, Zhang Y (2015) Calcitonin gene-related peptide can be selected as a predictive biomarker on progression and prognosis of knee osteoarthritis. Int Orthop 39:1237–1243PubMedCrossRef
18.
Zurück zum Zitat Lin WC, Yeh CH, Chien LC, Morone NE, Glick RM, Albers KM (2015) The anti-inflammatory actions of auricular point acupressure for chronic Low back pain. Evid Based Complement Alternat Med. doi:10.1155/2015/103570., Epub 2015 Jun 11 Lin WC, Yeh CH, Chien LC, Morone NE, Glick RM, Albers KM (2015) The anti-inflammatory actions of auricular point acupressure for chronic Low back pain. Evid Based Complement Alternat Med. doi:10.​1155/​2015/​103570.​, Epub 2015 Jun 11
19.
Zurück zum Zitat Carlsson CP, Sundler F, Wallengren J (2006) Cutaneous innervation before and after one treatment period of acupuncture. Br J Dermatol 155:970–6PubMedCrossRef Carlsson CP, Sundler F, Wallengren J (2006) Cutaneous innervation before and after one treatment period of acupuncture. Br J Dermatol 155:970–6PubMedCrossRef
20.
Zurück zum Zitat Saxler G, Löer F, Skumavc M, Pförtner J, Hanesch U (2007) Localization of SP- and CGRP-immunopositive nerve fibers in the hip joint of patients with painful osteoarthritis and of patients with painless failed total hip arthroplasties. Eur J Pain 11:67–74PubMedCrossRef Saxler G, Löer F, Skumavc M, Pförtner J, Hanesch U (2007) Localization of SP- and CGRP-immunopositive nerve fibers in the hip joint of patients with painful osteoarthritis and of patients with painless failed total hip arthroplasties. Eur J Pain 11:67–74PubMedCrossRef
21.
Zurück zum Zitat Wang H, Zhang X, He JY, Zheng XF, Li D, Li Z, Zhu JF, Shen C, Cai GQ, Chen XD (2015) Increasing expression of substance P and calcitonin gene-related peptide in synovial tissue and fluid contribute to the progress of arthritis in developmental dysplasia of the hip. Arthritis Res Ther 17:4PubMedPubMedCentralCrossRef Wang H, Zhang X, He JY, Zheng XF, Li D, Li Z, Zhu JF, Shen C, Cai GQ, Chen XD (2015) Increasing expression of substance P and calcitonin gene-related peptide in synovial tissue and fluid contribute to the progress of arthritis in developmental dysplasia of the hip. Arthritis Res Ther 17:4PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Takeshita M, Nakamura J, Ohtori S, Inoue G, Orita S, Miyagi M, Ishikawa T, Takahashi K (2012) Sensory innervation and inflammatory cytokines in hypertrophic synovia associated with pain transmission in osteoarthritis of the hip: a case–control study. Rheumatology (Oxford) 51:1790–5CrossRef Takeshita M, Nakamura J, Ohtori S, Inoue G, Orita S, Miyagi M, Ishikawa T, Takahashi K (2012) Sensory innervation and inflammatory cytokines in hypertrophic synovia associated with pain transmission in osteoarthritis of the hip: a case–control study. Rheumatology (Oxford) 51:1790–5CrossRef
23.
Zurück zum Zitat Sato J, Segami N, Kaneyama K, Yoshimura H, Fujimura K, Yoshitake Y (2004) Relationship of calcitonin gene-related peptide in synovial tissues and temporomandibular joint pain in humans. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 98:533–540PubMedCrossRef Sato J, Segami N, Kaneyama K, Yoshimura H, Fujimura K, Yoshitake Y (2004) Relationship of calcitonin gene-related peptide in synovial tissues and temporomandibular joint pain in humans. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 98:533–540PubMedCrossRef
24.
Zurück zum Zitat Ikeuchi M, Wang Q, Izumi M, Tani T (2012) Nociceptive sensory innervation of the posterior cruciate ligament in osteoarthritic knees. Arch Orthop Trauma Surg 132:891–895PubMedCrossRef Ikeuchi M, Wang Q, Izumi M, Tani T (2012) Nociceptive sensory innervation of the posterior cruciate ligament in osteoarthritic knees. Arch Orthop Trauma Surg 132:891–895PubMedCrossRef
25.
Zurück zum Zitat Lindh C, Liu Z, Welin M, Ordeberg G, Nyberg F (1999) Low calcitonin gene-related, peptide-like immunoreactivity in cerebrospinal fluid from chronic pain patients. Neuropeptides 33:517–521PubMedCrossRef Lindh C, Liu Z, Welin M, Ordeberg G, Nyberg F (1999) Low calcitonin gene-related, peptide-like immunoreactivity in cerebrospinal fluid from chronic pain patients. Neuropeptides 33:517–521PubMedCrossRef
26.
Zurück zum Zitat Brown MF, Hukkanen MV, McCarthy ID, Redfern DR, Batten JJ, Crock HV, Hughes SP, Polak JM (1997) Sensory and sympathetic innervation of the vertebral endplate in patients with degenerative disc disease. J Bone Joint Surg (Br) 79:147–153CrossRef Brown MF, Hukkanen MV, McCarthy ID, Redfern DR, Batten JJ, Crock HV, Hughes SP, Polak JM (1997) Sensory and sympathetic innervation of the vertebral endplate in patients with degenerative disc disease. J Bone Joint Surg (Br) 79:147–153CrossRef
27.
Zurück zum Zitat Ozawa T, Ohtori S, Inoue G, Aoki Y, Moriya H, Takahashi K (2006) The degenerated lumbar intervertebral disc is innervated primarily by peptide-containing sensory nerve fibers in humans. Spine (Phila Pa 1976) 31:2418–2422CrossRef Ozawa T, Ohtori S, Inoue G, Aoki Y, Moriya H, Takahashi K (2006) The degenerated lumbar intervertebral disc is innervated primarily by peptide-containing sensory nerve fibers in humans. Spine (Phila Pa 1976) 31:2418–2422CrossRef
28.
Zurück zum Zitat Alpar EK, Onuoha G, Killampalli VV, Waters R (2002) Management of chronic pain in whiplash injury. J Bone Joint Surg (Br) 84:807–811CrossRef Alpar EK, Onuoha G, Killampalli VV, Waters R (2002) Management of chronic pain in whiplash injury. J Bone Joint Surg (Br) 84:807–811CrossRef
29.
Zurück zum Zitat Takeuchi H, Kawaguchi S, Ohwada O, Kobayashi H, Hayakawa M, Takebayashi T, Troiqoe T, Sato N, Yamashita T (2007) Plasma neuropeptides in patients undergoing lumbar discectomy. Spine (Phila Pa 1976) 32:E79–84CrossRef Takeuchi H, Kawaguchi S, Ohwada O, Kobayashi H, Hayakawa M, Takebayashi T, Troiqoe T, Sato N, Yamashita T (2007) Plasma neuropeptides in patients undergoing lumbar discectomy. Spine (Phila Pa 1976) 32:E79–84CrossRef
30.
Zurück zum Zitat Onuoha GN, Alpar EK (1999) Calcitonin gene-related peptide and other neuropeptides in the plasma of patients with soft tissue injury. Life Sci 65:1351–1358PubMedCrossRef Onuoha GN, Alpar EK (1999) Calcitonin gene-related peptide and other neuropeptides in the plasma of patients with soft tissue injury. Life Sci 65:1351–1358PubMedCrossRef
31.
Zurück zum Zitat Larsson J, Ekblom A, Henriksson K, Lundeberg T, Theodorsson E (1991) Concentration of substance P, neurokinin A, calcitonin gene related peptide, neuropeptide Y and vasoactive intestinal polypeptide in synovial fluid from knee joints in patients suffering from rheumatoid arthritis. Scand J Rheumatol 20:326–335PubMedCrossRef Larsson J, Ekblom A, Henriksson K, Lundeberg T, Theodorsson E (1991) Concentration of substance P, neurokinin A, calcitonin gene related peptide, neuropeptide Y and vasoactive intestinal polypeptide in synovial fluid from knee joints in patients suffering from rheumatoid arthritis. Scand J Rheumatol 20:326–335PubMedCrossRef
32.
Zurück zum Zitat Bjur D, Alfredson H, Forsgren S (2005) The innervation pattern of the human Achilles tendon: studies of the normal and tendinosis tendon with markers for general and sensory innervation. Cell Tissue Res 320:201–206PubMedCrossRef Bjur D, Alfredson H, Forsgren S (2005) The innervation pattern of the human Achilles tendon: studies of the normal and tendinosis tendon with markers for general and sensory innervation. Cell Tissue Res 320:201–206PubMedCrossRef
33.
Zurück zum Zitat Danielson P, Andersson G, Alfredson H, Forsgren S (2008) Marked sympathetic component in the perivascular innervation of the dorsal paratendinous tissue of the patellar tendon in arthroscopically treated tendinosis patients. Knee Surg Sports Traumatol Arthrosc 16:621–626PubMedCrossRef Danielson P, Andersson G, Alfredson H, Forsgren S (2008) Marked sympathetic component in the perivascular innervation of the dorsal paratendinous tissue of the patellar tendon in arthroscopically treated tendinosis patients. Knee Surg Sports Traumatol Arthrosc 16:621–626PubMedCrossRef
34.
Zurück zum Zitat Sasaki K, Ohki G, Iba K, Kokai Y, Yamashita T, Wada T (2013) Innervation pattern at the undersurface of the extensor carpi radialis brevis tendon in recalcitrant tennis elbow. J Orthop Sci 18:528–535PubMedCrossRef Sasaki K, Ohki G, Iba K, Kokai Y, Yamashita T, Wada T (2013) Innervation pattern at the undersurface of the extensor carpi radialis brevis tendon in recalcitrant tennis elbow. J Orthop Sci 18:528–535PubMedCrossRef
35.
Zurück zum Zitat Samuelsson H, Ekman R, Hedner T (1993) CSF neuropeptides in cancer pain: effects of spinal opioid therapy. Acta Anaesthesiol Scand 37:502–508PubMedCrossRef Samuelsson H, Ekman R, Hedner T (1993) CSF neuropeptides in cancer pain: effects of spinal opioid therapy. Acta Anaesthesiol Scand 37:502–508PubMedCrossRef
36.
Zurück zum Zitat Jonhagen S, Ackermann P, Saartok T, Renstrom PA (2006) Calcitonin gene related peptide and neuropeptide Y in skeletal muscle after eccentric exercise: A microdialysis study. Br J Sports Med 40:264–267PubMedPubMedCentralCrossRef Jonhagen S, Ackermann P, Saartok T, Renstrom PA (2006) Calcitonin gene related peptide and neuropeptide Y in skeletal muscle after eccentric exercise: A microdialysis study. Br J Sports Med 40:264–267PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Barcena de Arellano ML, Arnold J, Vercellino GF, Chiantera V, Ebert AD, Schneider A, Mechsner S (2011) Influence of nerve growth factor in endometriosis-associated symptoms. Reprod Sci 18:1202–1210PubMedCrossRef Barcena de Arellano ML, Arnold J, Vercellino GF, Chiantera V, Ebert AD, Schneider A, Mechsner S (2011) Influence of nerve growth factor in endometriosis-associated symptoms. Reprod Sci 18:1202–1210PubMedCrossRef
38.
Zurück zum Zitat Tokushige N, Markham R, Russell P, Fraser IS (2006) Nerve fibres in peritoneal endometriosis. Hum Reprod 21:3001–3007PubMedCrossRef Tokushige N, Markham R, Russell P, Fraser IS (2006) Nerve fibres in peritoneal endometriosis. Hum Reprod 21:3001–3007PubMedCrossRef
39.
Zurück zum Zitat Tokushige N, Markham R, Russell P, Fraser IS (2007) Different types of small nerve fibers in eutopic endometrium and myometrium in women with endometriosis. Fertil Steril 88:795–803PubMedCrossRef Tokushige N, Markham R, Russell P, Fraser IS (2007) Different types of small nerve fibers in eutopic endometrium and myometrium in women with endometriosis. Fertil Steril 88:795–803PubMedCrossRef
40.
Zurück zum Zitat Tympanidis P, Terenghi G, Dowd P (2003) Increased innervation of the vulval vestibule in patients with vulvodynia. Br J Dermatol 148:1021–1027PubMedCrossRef Tympanidis P, Terenghi G, Dowd P (2003) Increased innervation of the vulval vestibule in patients with vulvodynia. Br J Dermatol 148:1021–1027PubMedCrossRef
41.
Zurück zum Zitat Yoshida N, Kuroda M, Suzuki T, Kamada K, Uchiyama K, Handa O, Takagi T, Yoshikawa T, Kuramoto H (2013) Role of nociceptors/neuropeptides in the pathogenesis of visceral hypersensitivity of nonerosive reflux disease. Dig Dis Sci 58:2237–2243PubMedCrossRef Yoshida N, Kuroda M, Suzuki T, Kamada K, Uchiyama K, Handa O, Takagi T, Yoshikawa T, Kuramoto H (2013) Role of nociceptors/neuropeptides in the pathogenesis of visceral hypersensitivity of nonerosive reflux disease. Dig Dis Sci 58:2237–2243PubMedCrossRef
42.
Zurück zum Zitat Mönnikes H, van der Voort IR, Wollenberg B, Heymann-Monnikes I, Tebbe JJ, Alt W, Arnold R, Klapp BF, Wiedenmann B, McGregor GP (2005) Gastric perception thresholds are low and sensory neuropeptide levels high in helicobacter pylori-positive functional dyspepsia. Digestion 71:111–123PubMedCrossRef Mönnikes H, van der Voort IR, Wollenberg B, Heymann-Monnikes I, Tebbe JJ, Alt W, Arnold R, Klapp BF, Wiedenmann B, McGregor GP (2005) Gastric perception thresholds are low and sensory neuropeptide levels high in helicobacter pylori-positive functional dyspepsia. Digestion 71:111–123PubMedCrossRef
43.
Zurück zum Zitat Buchler M, Weihe E, Friess H, Malfertheiner P, Bockman E, Müller S, Nohr D, Beger HG (1992) Changes in peptidergic innervation in chronic pancreatitis. Pancreas 7:183–192PubMedCrossRef Buchler M, Weihe E, Friess H, Malfertheiner P, Bockman E, Müller S, Nohr D, Beger HG (1992) Changes in peptidergic innervation in chronic pancreatitis. Pancreas 7:183–192PubMedCrossRef
44.
Zurück zum Zitat Währborg P, Eliasson T, Edvardsson N, Ekman R, Mannheimer C, Hedner T (1999) Acute ischemic chest pain is not associated with increased calcitonin gene-related peptide (CGRP) levels in peripheral plasma nor in the coronary circulation. Scand Cardiovasc J 33:295–299PubMedCrossRef Währborg P, Eliasson T, Edvardsson N, Ekman R, Mannheimer C, Hedner T (1999) Acute ischemic chest pain is not associated with increased calcitonin gene-related peptide (CGRP) levels in peripheral plasma nor in the coronary circulation. Scand Cardiovasc J 33:295–299PubMedCrossRef
45.
Zurück zum Zitat Geber C, Fondel R, Krämer HH, Rolke R, Treede RD, Sommer C, Birklein F (2007) Psychophysics, flare, and neurosecretory function in human pain models: capsaicin versus electrically evoked pain. J Pain 8:503–514PubMedCrossRef Geber C, Fondel R, Krämer HH, Rolke R, Treede RD, Sommer C, Birklein F (2007) Psychophysics, flare, and neurosecretory function in human pain models: capsaicin versus electrically evoked pain. J Pain 8:503–514PubMedCrossRef
46.
Zurück zum Zitat Schmelz M, Luz O, Averbeck B, Bickel A (1997) Plasma extravasation and neuropeptide release in human skin as measured by intradermal microdialysis. Neurosci Lett 230:117–120PubMedCrossRef Schmelz M, Luz O, Averbeck B, Bickel A (1997) Plasma extravasation and neuropeptide release in human skin as measured by intradermal microdialysis. Neurosci Lett 230:117–120PubMedCrossRef
47.
Zurück zum Zitat Simone DA, Nolano M, Johnson T, Wendelschafer-Crabb G, Kennedy WR (1998) Intradermal injection of capsaicin in humans produces degeneration and subsequent reinnervation of epidermal nerve fibers: Correlation with sensory function. J Neurosci 18:8947–8959PubMed Simone DA, Nolano M, Johnson T, Wendelschafer-Crabb G, Kennedy WR (1998) Intradermal injection of capsaicin in humans produces degeneration and subsequent reinnervation of epidermal nerve fibers: Correlation with sensory function. J Neurosci 18:8947–8959PubMed
48.
Zurück zum Zitat Krämer HH, Schmidt K, Leis S, Schmelz M, Sommer C, Birklein F (2005) Inhibition of neutral endopeptidase (NEP) facilitates neurogenic inflammation. Exp Neurol 195:179–184PubMedCrossRef Krämer HH, Schmidt K, Leis S, Schmelz M, Sommer C, Birklein F (2005) Inhibition of neutral endopeptidase (NEP) facilitates neurogenic inflammation. Exp Neurol 195:179–184PubMedCrossRef
49.
Zurück zum Zitat Couture R, Regoli D (1981) Inactivation of substance P and its C-terminal fragments in rat plasma and its inhibition by Captopril. Can J Physiol Pharmacol 59:621–625PubMedCrossRef Couture R, Regoli D (1981) Inactivation of substance P and its C-terminal fragments in rat plasma and its inhibition by Captopril. Can J Physiol Pharmacol 59:621–625PubMedCrossRef
50.
Zurück zum Zitat Hamed K, Giles N, Anderson J, Phillips JK, Dawson LF, Drummond P, Wallace H, Wood FM, Rea SM, Fear MW (2011) Changes in cutaneous innervation in patients with chronic pain after burns. Burns 37:631–637PubMedCrossRef Hamed K, Giles N, Anderson J, Phillips JK, Dawson LF, Drummond P, Wallace H, Wood FM, Rea SM, Fear MW (2011) Changes in cutaneous innervation in patients with chronic pain after burns. Burns 37:631–637PubMedCrossRef
51.
Zurück zum Zitat Kwak IS, Choi YH, Jang YC, Lee YK (2014) Immunohistochemical analysis of neuropeptides (protein gene product 9.5, substance P and calcitonin gene-related peptide) in hypertrophic burn scar with pain and itching. Burns 40:1661–1667PubMedCrossRef Kwak IS, Choi YH, Jang YC, Lee YK (2014) Immunohistochemical analysis of neuropeptides (protein gene product 9.5, substance P and calcitonin gene-related peptide) in hypertrophic burn scar with pain and itching. Burns 40:1661–1667PubMedCrossRef
52.
Zurück zum Zitat Onuoha GN, Alpar EK (2001) Levels of vasodilators (SP, CGRP) and vasoconstrictor (NPY) peptides in early human burns. Eur J Clin Invest 31:253–257PubMedCrossRef Onuoha GN, Alpar EK (2001) Levels of vasodilators (SP, CGRP) and vasoconstrictor (NPY) peptides in early human burns. Eur J Clin Invest 31:253–257PubMedCrossRef
53.
Zurück zum Zitat Salomon J, Baran E (2008) The role of selected neuropeptides in pathogenesis of atopic dermatitis. J Eur Acad Dermatol Venereol 22:223–228PubMed Salomon J, Baran E (2008) The role of selected neuropeptides in pathogenesis of atopic dermatitis. J Eur Acad Dermatol Venereol 22:223–228PubMed
54.
Zurück zum Zitat Ringkamp M, Schepers RJ, Shimada SG, Johanek LM, Hartke TV, Borzan J, Shim B, LaMotte RH, Meyer RA (2011) A role for nociceptive, myelinated nerve fibers in itch sensation. J Neurosci 31:14841–14849PubMedPubMedCentralCrossRef Ringkamp M, Schepers RJ, Shimada SG, Johanek LM, Hartke TV, Borzan J, Shim B, LaMotte RH, Meyer RA (2011) A role for nociceptive, myelinated nerve fibers in itch sensation. J Neurosci 31:14841–14849PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Birklein F, Schmelz M, Schifter S, Weber M (2001) The important role of neuropeptides in complex regional pain syndrome. Neurology 57:2179–2184PubMedCrossRef Birklein F, Schmelz M, Schifter S, Weber M (2001) The important role of neuropeptides in complex regional pain syndrome. Neurology 57:2179–2184PubMedCrossRef
56.
Zurück zum Zitat Schinkel C, Scherens A, Köller M, Roellecke G, Muhr G, Maier C (2009) Systemic inflammatory mediators in post-traumatic complex regional pain syndrome (CRPS I) - longitudinal investigations and differences to control groups. Eur J Med Res 14:130–135PubMedPubMedCentralCrossRef Schinkel C, Scherens A, Köller M, Roellecke G, Muhr G, Maier C (2009) Systemic inflammatory mediators in post-traumatic complex regional pain syndrome (CRPS I) - longitudinal investigations and differences to control groups. Eur J Med Res 14:130–135PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Albrecht PJ, Hines S, Eisenberg E, Pud D, Finlay DR, Connolly MK, Paré M, Davar G, Rice FL (2006) Pathologic alterations of cutaneous innervation and vasculature in affected limbs from patients with complex regional pain syndrome. Pain 120:244–266PubMedCrossRef Albrecht PJ, Hines S, Eisenberg E, Pud D, Finlay DR, Connolly MK, Paré M, Davar G, Rice FL (2006) Pathologic alterations of cutaneous innervation and vasculature in affected limbs from patients with complex regional pain syndrome. Pain 120:244–266PubMedCrossRef
58.
Zurück zum Zitat Hou Q, Barr T, Gee L, Vickers J, Wymer J, Borsani E, Rodella L, Getsios S, Burdo T, Eisenberg E, Guha U, Lavker R, Kessler J, Chittur S, Fiorino D, Rice F, Albrecht P (2011) Keratinocyte expression of calcitonin gene-related peptide β: implications for neuropathic and inflammatory pain mechanisms. Pain 152:2036–2051PubMedPubMedCentralCrossRef Hou Q, Barr T, Gee L, Vickers J, Wymer J, Borsani E, Rodella L, Getsios S, Burdo T, Eisenberg E, Guha U, Lavker R, Kessler J, Chittur S, Fiorino D, Rice F, Albrecht P (2011) Keratinocyte expression of calcitonin gene-related peptide β: implications for neuropathic and inflammatory pain mechanisms. Pain 152:2036–2051PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Kalliomäki M, Kieseritzky JV, Schmidt R, Hägglöf B, Karlsten R, Sjögren N, Albrecht P, Gee L, Rice F, Wiig M, Schmelz M, Gordh T (2011) Structural and functional differences between neuropathy with and without pain? Exp Neurol 231:199–206PubMedCrossRef Kalliomäki M, Kieseritzky JV, Schmidt R, Hägglöf B, Karlsten R, Sjögren N, Albrecht P, Gee L, Rice F, Wiig M, Schmelz M, Gordh T (2011) Structural and functional differences between neuropathy with and without pain? Exp Neurol 231:199–206PubMedCrossRef
60.
Zurück zum Zitat Lindqvist A, Rivero-Melian C, Turan I, Fried K (2000) Neuropeptide- and tyrosine hydroxylase-immunoreactive nerve fibers in painful Morton’s neuromas. Muscle Nerve 23:1214–1218PubMedCrossRef Lindqvist A, Rivero-Melian C, Turan I, Fried K (2000) Neuropeptide- and tyrosine hydroxylase-immunoreactive nerve fibers in painful Morton’s neuromas. Muscle Nerve 23:1214–1218PubMedCrossRef
61.
Zurück zum Zitat Awawdeh LA, Lundy FT, Linden GJ, Shaw KJG, Lamey PJ (2002) Quantitative analysis of substance P, neurokinin A and calcitonin gene-related peptide in gingival crevicular fluid associated with painful human teeth. Eur J Oral Sci 110:185–191PubMedCrossRef Awawdeh LA, Lundy FT, Linden GJ, Shaw KJG, Lamey PJ (2002) Quantitative analysis of substance P, neurokinin A and calcitonin gene-related peptide in gingival crevicular fluid associated with painful human teeth. Eur J Oral Sci 110:185–191PubMedCrossRef
62.
Zurück zum Zitat Zidverc-Trajkovic J, Stanimirovic D, Obrenovic R, Tajti J, Vécsei K, Gardi J, Németh J, Mijajlovic M, Sternic N, Jankovic L (2009) Calcitonin gene-related peptide levels in saliva of patients with burning mouth syndrome. J Oral Pathol Med 38:29–33PubMedCrossRef Zidverc-Trajkovic J, Stanimirovic D, Obrenovic R, Tajti J, Vécsei K, Gardi J, Németh J, Mijajlovic M, Sternic N, Jankovic L (2009) Calcitonin gene-related peptide levels in saliva of patients with burning mouth syndrome. J Oral Pathol Med 38:29–33PubMedCrossRef
63.
Zurück zum Zitat Boras VV, Savage NW, Brailo V, Lukac J, Lukac M, Alajbeg IZ (2010) Salivary and serum levels of substance P, neurokinin A and calcitonin gene related peptide in burning mouth syndrome. Med Oral Patol Oral Cir Bucal 15:e427–431PubMedCrossRef Boras VV, Savage NW, Brailo V, Lukac J, Lukac M, Alajbeg IZ (2010) Salivary and serum levels of substance P, neurokinin A and calcitonin gene related peptide in burning mouth syndrome. Med Oral Patol Oral Cir Bucal 15:e427–431PubMedCrossRef
64.
Zurück zum Zitat Chavarría-Bolaños D, Martinez-Zumaran A, Lombana N, Flores-Reyes H, Pozos-Guillen A (2014) Expression of substance P, calcitonin gene-related peptide, β-endorphin and methionine-enkephalin in human dental pulp tissue after orthodontic intrusion A pilot study. Angle Orthod 84:521–526PubMedCrossRef Chavarría-Bolaños D, Martinez-Zumaran A, Lombana N, Flores-Reyes H, Pozos-Guillen A (2014) Expression of substance P, calcitonin gene-related peptide, β-endorphin and methionine-enkephalin in human dental pulp tissue after orthodontic intrusion A pilot study. Angle Orthod 84:521–526PubMedCrossRef
65.
Zurück zum Zitat Attal N, de Andrade DC, Adam F, Ranoux D, Teixeira MJ, Galhardoni R, Raicher I, Üceyler N, Sommer C, Bouhassira D (2016) Safety and efficacy of repeated injections of botulinum toxin A in peripheral neuropathic pain (BOTNEP): A randomised, double-blind, placebo-controlled trial. Lancet Neurol 15:555–565PubMedCrossRef Attal N, de Andrade DC, Adam F, Ranoux D, Teixeira MJ, Galhardoni R, Raicher I, Üceyler N, Sommer C, Bouhassira D (2016) Safety and efficacy of repeated injections of botulinum toxin A in peripheral neuropathic pain (BOTNEP): A randomised, double-blind, placebo-controlled trial. Lancet Neurol 15:555–565PubMedCrossRef
68.
Zurück zum Zitat Jin Y, Smith C, Monteith D, Brown R, Camporeale A, McNearney T, Deeg M, Raddad E, de la Pena A, Kivitz A, Schnitzer T (2016) LY2951742, a monoclonal antibody against CGRP, failed to reduce signs and symptoms of knee osteoarthritis. Osteoarthritis Cartilage 24:S50CrossRef Jin Y, Smith C, Monteith D, Brown R, Camporeale A, McNearney T, Deeg M, Raddad E, de la Pena A, Kivitz A, Schnitzer T (2016) LY2951742, a monoclonal antibody against CGRP, failed to reduce signs and symptoms of knee osteoarthritis. Osteoarthritis Cartilage 24:S50CrossRef
70.
Zurück zum Zitat Tajti J, Uddman R, Edvinsson L (2001) Neuropeptide localization in the ‘migraine generator’ region of the human brainstem. Cephalalgia 21:96–101PubMedCrossRef Tajti J, Uddman R, Edvinsson L (2001) Neuropeptide localization in the ‘migraine generator’ region of the human brainstem. Cephalalgia 21:96–101PubMedCrossRef
71.
Zurück zum Zitat van Rossum D, Hanisch UK, Quirion R (1997) Neuroanatomical localization, pharmacological characterization and functions of CGRP, related peptides and their receptors. Neurosci Biobehav Rev 21:649–678PubMedCrossRef van Rossum D, Hanisch UK, Quirion R (1997) Neuroanatomical localization, pharmacological characterization and functions of CGRP, related peptides and their receptors. Neurosci Biobehav Rev 21:649–678PubMedCrossRef
72.
Zurück zum Zitat Chakravarty P, Suthar TP, Coppock HA, Nicholl CG, Bloom SR, Legon S, Smith DM (2000) CGRP and adrenomedullin binding correlates with transcript levels for calcitonin receptor-like receptor (CRLR) and receptor activity modifying proteins (RAMPs) in rat tissues. BrJ Pharmacol 130:189–195CrossRef Chakravarty P, Suthar TP, Coppock HA, Nicholl CG, Bloom SR, Legon S, Smith DM (2000) CGRP and adrenomedullin binding correlates with transcript levels for calcitonin receptor-like receptor (CRLR) and receptor activity modifying proteins (RAMPs) in rat tissues. BrJ Pharmacol 130:189–195CrossRef
73.
Zurück zum Zitat Ma W, Chabot JG, Powell KJ, Jhamandas K, Dickerson IM, Quirion R (2003) Localization and modulation of calcitonin gene-related peptide-receptor component protein-immunoreactive cells in the rat central and peripheral nervous systems. Neuroscience 120:677–694PubMedCrossRef Ma W, Chabot JG, Powell KJ, Jhamandas K, Dickerson IM, Quirion R (2003) Localization and modulation of calcitonin gene-related peptide-receptor component protein-immunoreactive cells in the rat central and peripheral nervous systems. Neuroscience 120:677–694PubMedCrossRef
74.
Zurück zum Zitat Cottrell GS, Alemi F, Kirkland JG, Grady EF, Corvera CT, Bhargava A (2012) Localization of calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein 1 (RAMP1) in human gastrointestinal tract. Peptides 35:202–211PubMedPubMedCentralCrossRef Cottrell GS, Alemi F, Kirkland JG, Grady EF, Corvera CT, Bhargava A (2012) Localization of calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein 1 (RAMP1) in human gastrointestinal tract. Peptides 35:202–211PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Cottrell GS, Roosterman D, Marvizon JC, Song B, Wick E, Pikios S, Wong H, Berthelier C, Tang Y, Sternini C, Bunnett NW, Grady EF (2005) Localization of calcitonin receptor-like receptor and receptor activity modifying protein 1 in enteric neurons, dorsal root ganglia, and the spinal cord of the rat. J Comp Neurol 490:239–255PubMedCrossRef Cottrell GS, Roosterman D, Marvizon JC, Song B, Wick E, Pikios S, Wong H, Berthelier C, Tang Y, Sternini C, Bunnett NW, Grady EF (2005) Localization of calcitonin receptor-like receptor and receptor activity modifying protein 1 in enteric neurons, dorsal root ganglia, and the spinal cord of the rat. J Comp Neurol 490:239–255PubMedCrossRef
76.
Zurück zum Zitat Pokabla MJ, Dickerson IM, Papka RE (2002) Calcitonin gene-related peptide-receptor component protein expression in the uterine cervix, lumbosacral spinal cord, and dorsal root ganglia. Peptides 23:507–514PubMedCrossRef Pokabla MJ, Dickerson IM, Papka RE (2002) Calcitonin gene-related peptide-receptor component protein expression in the uterine cervix, lumbosacral spinal cord, and dorsal root ganglia. Peptides 23:507–514PubMedCrossRef
77.
Zurück zum Zitat McCarthy PW, Lawson SN (1990) Cell type and conduction velocity of rat primary sensory neurons with calcitonin gene-related peptide-like immunoreactivity. Neuroscience 34:623–632PubMedCrossRef McCarthy PW, Lawson SN (1990) Cell type and conduction velocity of rat primary sensory neurons with calcitonin gene-related peptide-like immunoreactivity. Neuroscience 34:623–632PubMedCrossRef
78.
Zurück zum Zitat Belanger S, Ma W, Chabot JG, Quirion R (2002) Expression of calcitonin gene-related peptide, substance P and protein kinase C in cultured dorsal root ganglion neurons following chronic exposure to mu, delta and kappa opiates. Neuroscience 115:441–453PubMedCrossRef Belanger S, Ma W, Chabot JG, Quirion R (2002) Expression of calcitonin gene-related peptide, substance P and protein kinase C in cultured dorsal root ganglion neurons following chronic exposure to mu, delta and kappa opiates. Neuroscience 115:441–453PubMedCrossRef
79.
Zurück zum Zitat Yang YL, Yao KH, Li ZW (2003) Similarities of SP-, NKA- and NKB-induced currents in rat dorsal root ganglion neurons. Brain Res 991:18–25PubMedCrossRef Yang YL, Yao KH, Li ZW (2003) Similarities of SP-, NKA- and NKB-induced currents in rat dorsal root ganglion neurons. Brain Res 991:18–25PubMedCrossRef
80.
Zurück zum Zitat Gibson SJ, Polak JM, Bloom SR, Sabate IM, Mulderry PM, Ghatei MA, McGregor GP, Morrison JF, Kelly JS, Evans RM (1984) Calcitonin gene-related peptide immunoreactivity in the spinal cord of man and of eight other species. J Neurosci 4:3101–3111PubMed Gibson SJ, Polak JM, Bloom SR, Sabate IM, Mulderry PM, Ghatei MA, McGregor GP, Morrison JF, Kelly JS, Evans RM (1984) Calcitonin gene-related peptide immunoreactivity in the spinal cord of man and of eight other species. J Neurosci 4:3101–3111PubMed
81.
Zurück zum Zitat Fernihough J, Gentry C, Bevan S, Winter J (2005) Regulation of calcitonin gene-related peptide and TRPV1 in a rat model of osteoarthritis. Neurosci Lett 388:75–80PubMedCrossRef Fernihough J, Gentry C, Bevan S, Winter J (2005) Regulation of calcitonin gene-related peptide and TRPV1 in a rat model of osteoarthritis. Neurosci Lett 388:75–80PubMedCrossRef
82.
Zurück zum Zitat Morion CR, Hutchison WD (1989) Release of sensory neuropeptides in the spinal cord: Studies with calcitonin gene-related peptide and galanin. Neuroscience 31:807–815CrossRef Morion CR, Hutchison WD (1989) Release of sensory neuropeptides in the spinal cord: Studies with calcitonin gene-related peptide and galanin. Neuroscience 31:807–815CrossRef
83.
Zurück zum Zitat Sauer SK, Bove GM, Averbeck B, Reeh PW (1999) Rat peripheral nerve components release calcitonin gene-related peptide and prostaglandin E2 in response to noxious stimuli: Evidence that nervi nervorum are nociceptors. Neuroscience 92:319–325PubMedCrossRef Sauer SK, Bove GM, Averbeck B, Reeh PW (1999) Rat peripheral nerve components release calcitonin gene-related peptide and prostaglandin E2 in response to noxious stimuli: Evidence that nervi nervorum are nociceptors. Neuroscience 92:319–325PubMedCrossRef
84.
Zurück zum Zitat Schaible HG, Freudenberger U, Neugebauer V, Stiller RU (1994) Intraspinal release of immunoreactive calcitonin gene-related peptide during development of inflammation in the joint in vivo-a study with antibody microprobes in cat and rat. Neuroscience 62:1293–1305PubMedCrossRef Schaible HG, Freudenberger U, Neugebauer V, Stiller RU (1994) Intraspinal release of immunoreactive calcitonin gene-related peptide during development of inflammation in the joint in vivo-a study with antibody microprobes in cat and rat. Neuroscience 62:1293–1305PubMedCrossRef
85.
Zurück zum Zitat Ogbonna AC, Clark AK, Gentry C, Hobbs C, Malcangio M (2013) Pain-like behaviour and spinal changes in the monosodium iodoacetate model of osteoarthritis in C57Bl/6 mice. Eur J Pain 17:514–526PubMedCrossRef Ogbonna AC, Clark AK, Gentry C, Hobbs C, Malcangio M (2013) Pain-like behaviour and spinal changes in the monosodium iodoacetate model of osteoarthritis in C57Bl/6 mice. Eur J Pain 17:514–526PubMedCrossRef
86.
Zurück zum Zitat Wimalawansa SJ, MacIntyre I (1988) Heterogeneity of plasma calcitonin gene-related peptide: partial characterisation of immunoreactive forms. Peptides 9:407–410PubMedCrossRef Wimalawansa SJ, MacIntyre I (1988) Heterogeneity of plasma calcitonin gene-related peptide: partial characterisation of immunoreactive forms. Peptides 9:407–410PubMedCrossRef
87.
Zurück zum Zitat Zaidi M, Bevis PJ, Abeyasekera G, Girgis SI, Wimalawansa SJ, Morris HR, MacIntyre I (1986) The origin of circulating calcitonin gene-related peptide in the rat. J Endocrinol 110:185–190PubMedCrossRef Zaidi M, Bevis PJ, Abeyasekera G, Girgis SI, Wimalawansa SJ, Morris HR, MacIntyre I (1986) The origin of circulating calcitonin gene-related peptide in the rat. J Endocrinol 110:185–190PubMedCrossRef
88.
Zurück zum Zitat Buzzi MG, Carter WB, Shimizu T, Health H 3rd, Moskowitz MA (1991) Dihydroergotamine and sumatriptan attenuate levels of CGRP in plasma in rat superior sagittal sinus during electrical stimulation of the trigeminal ganglion. Neuropharmacology 30:1193–1200PubMedCrossRef Buzzi MG, Carter WB, Shimizu T, Health H 3rd, Moskowitz MA (1991) Dihydroergotamine and sumatriptan attenuate levels of CGRP in plasma in rat superior sagittal sinus during electrical stimulation of the trigeminal ganglion. Neuropharmacology 30:1193–1200PubMedCrossRef
89.
Zurück zum Zitat Levine JD, Fields HL, Basbaum AI (1993) Peptides and the primary afferent nociceptor. J Neurosci 13:2273–2286PubMed Levine JD, Fields HL, Basbaum AI (1993) Peptides and the primary afferent nociceptor. J Neurosci 13:2273–2286PubMed
90.
Zurück zum Zitat Morton CR, Hutchison WD (1990) Morphine does not reduce the intraspinal release of calcitonin gene-related peptide in the cat. Neurosci Lett 117:319–324PubMedCrossRef Morton CR, Hutchison WD (1990) Morphine does not reduce the intraspinal release of calcitonin gene-related peptide in the cat. Neurosci Lett 117:319–324PubMedCrossRef
91.
Zurück zum Zitat Weihe E, Schäfer MK, Nohr D, Persson S (1994) Expression of neuropeptides, neuropeptide receptors and neuropeptide processing enzymes in spinal neurons and peripheral non-neural cells and plasticity in models of inflammatory pain. In: Hokfelt T, Schaible HG, Schmidt RF (eds) Neuropeptides, nociception and Pain. Chapman & Hall, London Weihe E, Schäfer MK, Nohr D, Persson S (1994) Expression of neuropeptides, neuropeptide receptors and neuropeptide processing enzymes in spinal neurons and peripheral non-neural cells and plasticity in models of inflammatory pain. In: Hokfelt T, Schaible HG, Schmidt RF (eds) Neuropeptides, nociception and Pain. Chapman & Hall, London
92.
Zurück zum Zitat Leem JW, Gwak YS, Lee EH, Chung SS, Ys K, Nam TS (2001) Effects of iontophoretically applied substance P, calcitonin gene-related peptide on excitability of dorsal horn neurones in rats. Yonsei Med J 42:74–83PubMedCrossRef Leem JW, Gwak YS, Lee EH, Chung SS, Ys K, Nam TS (2001) Effects of iontophoretically applied substance P, calcitonin gene-related peptide on excitability of dorsal horn neurones in rats. Yonsei Med J 42:74–83PubMedCrossRef
93.
Zurück zum Zitat Sun RQ, Tu YJ, Lawand NB, Yan JY, Lin Q, Willis WD (2004) Calcitonin gene-related peptide receptor activation produces PKA- and PKC-dependent mechanical hyperalgesia and central sensitization. J Neurophysiol 92:2859–2866PubMedCrossRef Sun RQ, Tu YJ, Lawand NB, Yan JY, Lin Q, Willis WD (2004) Calcitonin gene-related peptide receptor activation produces PKA- and PKC-dependent mechanical hyperalgesia and central sensitization. J Neurophysiol 92:2859–2866PubMedCrossRef
94.
Zurück zum Zitat Kessler F, Habelt C, Averbeck B, Reeh PW, Kress M (1999) Heat-induced release of CGRP from isolated rat skin and effects of bradykinin and the protein kinase C activator PMA. Pain 83:289–295PubMedCrossRef Kessler F, Habelt C, Averbeck B, Reeh PW, Kress M (1999) Heat-induced release of CGRP from isolated rat skin and effects of bradykinin and the protein kinase C activator PMA. Pain 83:289–295PubMedCrossRef
95.
Zurück zum Zitat Segond von Banchet G, Pastor A, Biskup C, Schlegel C, Benndorf K, Schaible HG (2002) Localization of functional calcitonin gene-related peptide binding sites in a subpopulation of cultured dorsal root ganglion neurons. Neuroscience 110:131–145PubMedCrossRef Segond von Banchet G, Pastor A, Biskup C, Schlegel C, Benndorf K, Schaible HG (2002) Localization of functional calcitonin gene-related peptide binding sites in a subpopulation of cultured dorsal root ganglion neurons. Neuroscience 110:131–145PubMedCrossRef
96.
Zurück zum Zitat Natura G, von Banchet GS, Schaible HG (2005) Calcitonin gene-related peptide enhances TTX-resistant sodium currents in cultured dorsal root ganglion neurons from adult rats. Pain 116:194–204PubMedCrossRef Natura G, von Banchet GS, Schaible HG (2005) Calcitonin gene-related peptide enhances TTX-resistant sodium currents in cultured dorsal root ganglion neurons from adult rats. Pain 116:194–204PubMedCrossRef
97.
Zurück zum Zitat Shi X, Wang L, Li X, Sahbaie P, Kingery WS, Clark JD (2011) Neuropeptides contribute to peripheral nociceptive sensitization by regulating interleukin-1β production in keratinocytes. Anesth Analg 113:175–183PubMedPubMedCentralCrossRef Shi X, Wang L, Li X, Sahbaie P, Kingery WS, Clark JD (2011) Neuropeptides contribute to peripheral nociceptive sensitization by regulating interleukin-1β production in keratinocytes. Anesth Analg 113:175–183PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Pedersen-Bjergaard U, Nielsen LB, Jensen K, Edvinsson L, Jansen I, Olesen J (1991) Calcitonin gene-related peptide, neurokinin A and substance P: Effects on Nociception and neurogenic inflammation in human skin and temporal muscle. Peptides 12:333–337PubMedCrossRef Pedersen-Bjergaard U, Nielsen LB, Jensen K, Edvinsson L, Jansen I, Olesen J (1991) Calcitonin gene-related peptide, neurokinin A and substance P: Effects on Nociception and neurogenic inflammation in human skin and temporal muscle. Peptides 12:333–337PubMedCrossRef
99.
Zurück zum Zitat Pereira da Silva JA, Carmo-Fonseca M (1990) Peptide containing nerves in human synovium: Immunohistochemical evidence for decreased innervation in rheumatoid arthritis. J Rheumatol 17:1592–1599PubMed Pereira da Silva JA, Carmo-Fonseca M (1990) Peptide containing nerves in human synovium: Immunohistochemical evidence for decreased innervation in rheumatoid arthritis. J Rheumatol 17:1592–1599PubMed
100.
Zurück zum Zitat Gibbins IL, Wattchow D, Coventry B (1987) Two immunohistochemically identified populations of calcitonin gene-related peptide (CGRP)-immunoreactive axons in human skin. Brain Res 414:143–148PubMedCrossRef Gibbins IL, Wattchow D, Coventry B (1987) Two immunohistochemically identified populations of calcitonin gene-related peptide (CGRP)-immunoreactive axons in human skin. Brain Res 414:143–148PubMedCrossRef
101.
Zurück zum Zitat Fujimori A, Saito A, Kimura S, Watanabe T, Uchiyama Y, Kawasaki H, Goto K (1989) Neurogenic vasodilation and release of calcitonin gene-related peptide (CGRP) from perivascular nerves in the rat mesenteric artery. Biochem Biophys Res Commun 165:1391–1398PubMedCrossRef Fujimori A, Saito A, Kimura S, Watanabe T, Uchiyama Y, Kawasaki H, Goto K (1989) Neurogenic vasodilation and release of calcitonin gene-related peptide (CGRP) from perivascular nerves in the rat mesenteric artery. Biochem Biophys Res Commun 165:1391–1398PubMedCrossRef
102.
Zurück zum Zitat Uddman R, Edvinsson L, Ekblad E, Håkanson R, Sundler F (1986) Calcitonin gene-related peptide (CGRP): perivascular distribution and vasodilatory effects. Regul Pept 15:1–23PubMedCrossRef Uddman R, Edvinsson L, Ekblad E, Håkanson R, Sundler F (1986) Calcitonin gene-related peptide (CGRP): perivascular distribution and vasodilatory effects. Regul Pept 15:1–23PubMedCrossRef
103.
Zurück zum Zitat Lee JK, Jung JS, Park SH, Sim YB, Sub HW (2013) Deficiency of alpha-calcitonin gene-related peptide induces inflammatory responses and lethality in sepsis. Cytokine 64:548–554PubMedCrossRef Lee JK, Jung JS, Park SH, Sim YB, Sub HW (2013) Deficiency of alpha-calcitonin gene-related peptide induces inflammatory responses and lethality in sepsis. Cytokine 64:548–554PubMedCrossRef
104.
Zurück zum Zitat Ishida K, Kawamata T, Tanaka S, Shindo T, Kawamata M (2014) Calcitonin gene-related peptide is involved in inflammatory pain but not in postoperative pain. Anesthesiology 121:1068–1079PubMedCrossRef Ishida K, Kawamata T, Tanaka S, Shindo T, Kawamata M (2014) Calcitonin gene-related peptide is involved in inflammatory pain but not in postoperative pain. Anesthesiology 121:1068–1079PubMedCrossRef
105.
Zurück zum Zitat Nakamura-Craig M, Gill BK (1991) Effect of neurokinin A, substance P and calcitonin gene related peptide in peripheral hyperalgesia in the rat paw. Neurosci Lett 124:49–51PubMedCrossRef Nakamura-Craig M, Gill BK (1991) Effect of neurokinin A, substance P and calcitonin gene related peptide in peripheral hyperalgesia in the rat paw. Neurosci Lett 124:49–51PubMedCrossRef
106.
Zurück zum Zitat Schaible HG, Schmidt RF (1988) Excitation and sensitization of fine articular afferents from cat’s knee joint by prostaglandin E2. J Physiol 403:91–104PubMedPubMedCentralCrossRef Schaible HG, Schmidt RF (1988) Excitation and sensitization of fine articular afferents from cat’s knee joint by prostaglandin E2. J Physiol 403:91–104PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Birrell GJ, McQueen DS, Iggo A, Coleman RA, Grubb BD (1991) PGI2-induced activation and sensitization of articular mechanonociceptors. Neurosci Lett 124:5–8PubMedCrossRef Birrell GJ, McQueen DS, Iggo A, Coleman RA, Grubb BD (1991) PGI2-induced activation and sensitization of articular mechanonociceptors. Neurosci Lett 124:5–8PubMedCrossRef
108.
Zurück zum Zitat Wang H, Ehnert C, Brenner GJ, Woolf CJ (2006) Bradykinin and peripheral sensitization. Biol Chem 387:11–14PubMed Wang H, Ehnert C, Brenner GJ, Woolf CJ (2006) Bradykinin and peripheral sensitization. Biol Chem 387:11–14PubMed
109.
Zurück zum Zitat Bullock CM, Kelly S (2013) Calcitonin gene-related peptide receptor antagonists: beyond migraine pain--a possible analgesic strategy for osteoarthritis? Curr Pain Headache Rep 17:375PubMedPubMedCentralCrossRef Bullock CM, Kelly S (2013) Calcitonin gene-related peptide receptor antagonists: beyond migraine pain--a possible analgesic strategy for osteoarthritis? Curr Pain Headache Rep 17:375PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Haegerstrand A, Dalsgaard CJ, Jonzon B, Larsson O, Nilsson J (1990) Calcitonin gene-related peptide stimulates proliferation of human endothelial cells. Proc Natl Acad Sci U S A 87:3299–3303PubMedPubMedCentralCrossRef Haegerstrand A, Dalsgaard CJ, Jonzon B, Larsson O, Nilsson J (1990) Calcitonin gene-related peptide stimulates proliferation of human endothelial cells. Proc Natl Acad Sci U S A 87:3299–3303PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Arendt-Nielsen L, Nie H, Laursen MB, Laursen BS, Madeleine P, Simonsen OH, Graven-Nielsen T (2010) Sensitization in patients with painful knee osteoarthrosis. Pain 149:573–581PubMedCrossRef Arendt-Nielsen L, Nie H, Laursen MB, Laursen BS, Madeleine P, Simonsen OH, Graven-Nielsen T (2010) Sensitization in patients with painful knee osteoarthrosis. Pain 149:573–581PubMedCrossRef
112.
Zurück zum Zitat Axelsson HE, Minde JK, Sonesson A, Toolanen G, Högestätt ED, Zygmunt PM (2009) Transient receptor potential vanilloid 1, vanilloid 2 and melastatin 8 immunoreactive nerve fibers in human skin from individuals with and without Norrbottnian congenital insensitivity to pain. Neuroscience 162:1322–1332PubMedCrossRef Axelsson HE, Minde JK, Sonesson A, Toolanen G, Högestätt ED, Zygmunt PM (2009) Transient receptor potential vanilloid 1, vanilloid 2 and melastatin 8 immunoreactive nerve fibers in human skin from individuals with and without Norrbottnian congenital insensitivity to pain. Neuroscience 162:1322–1332PubMedCrossRef
113.
Zurück zum Zitat Sulaiman H, Gabella G, Davis MSc C, Se M, Boulos P, Laurent GJ, Herrick SE (2001) Presence and distribution of sensory nerve fibers in human peritoneal adhesions. Ann Surg 234:256–261PubMedPubMedCentralCrossRef Sulaiman H, Gabella G, Davis MSc C, Se M, Boulos P, Laurent GJ, Herrick SE (2001) Presence and distribution of sensory nerve fibers in human peritoneal adhesions. Ann Surg 234:256–261PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Mapp PI, McWilliams DF, Turley MJ, Hargin E, Walsh DA (2012) A role for the sensory neuropeptide calcitonin gene-related peptide in endothelial cell proliferation in vivo. Br J Pharmacol 166:1261–1271PubMedPubMedCentralCrossRef Mapp PI, McWilliams DF, Turley MJ, Hargin E, Walsh DA (2012) A role for the sensory neuropeptide calcitonin gene-related peptide in endothelial cell proliferation in vivo. Br J Pharmacol 166:1261–1271PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Amann R (1990) Desensitization of capsaicin-evoked neuropeptide release—influence of Ca2+ and temperature. Naunyn Schmiedebergs Arch Pharmacol 342:671–676PubMedCrossRef Amann R (1990) Desensitization of capsaicin-evoked neuropeptide release—influence of Ca2+ and temperature. Naunyn Schmiedebergs Arch Pharmacol 342:671–676PubMedCrossRef
116.
Zurück zum Zitat Van der Schueren BJ1, Rogiers A, Vanmolkot FH, Van Hecken A, Depré M, Kane SA, De Lepeleire I, Sinclair SR, de Hoon JN (2008) Calcitonin gene-related peptide8-37 antagonizes capsaicin-induced vasodilation in the skin: evaluation of a human in vivo pharmacodynamic model. J Pharmacol Exp Ther 325:248-255. Van der Schueren BJ1, Rogiers A, Vanmolkot FH, Van Hecken A, Depré M, Kane SA, De Lepeleire I, Sinclair SR, de Hoon JN (2008) Calcitonin gene-related peptide8-37 antagonizes capsaicin-induced vasodilation in the skin: evaluation of a human in vivo pharmacodynamic model. J Pharmacol Exp Ther 325:248-255.
117.
Zurück zum Zitat Li CC, Vermeersch S, Denney WS, Kennedy WP, Palcza J, Gipson A, Han TH, Blanchard R, De Lepeleire I, Depré M, Murphy MG, Van Dyck K, de Hoon JN (2015) Characterizing the PK/PD relationship for inhibition of capsaicin-induced dermal vasodilatation by MK-3207, an oral calcitonin gene related peptide receptor antagonist. Br J Clin Pharmacol 79:831–837PubMedPubMedCentralCrossRef Li CC, Vermeersch S, Denney WS, Kennedy WP, Palcza J, Gipson A, Han TH, Blanchard R, De Lepeleire I, Depré M, Murphy MG, Van Dyck K, de Hoon JN (2015) Characterizing the PK/PD relationship for inhibition of capsaicin-induced dermal vasodilatation by MK-3207, an oral calcitonin gene related peptide receptor antagonist. Br J Clin Pharmacol 79:831–837PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Sinclair SR, Kane SA, Van der Schueren BJ, Xiao A, Willson KJ, Boyle J (2010) Inhibition of capsaicin-induced increase in dermal blood flow by the oral CGRP receptor antagonist, telcagepant (MK-0974). Br J Clin Pharmacol 69:15–22PubMedPubMedCentralCrossRef Sinclair SR, Kane SA, Van der Schueren BJ, Xiao A, Willson KJ, Boyle J (2010) Inhibition of capsaicin-induced increase in dermal blood flow by the oral CGRP receptor antagonist, telcagepant (MK-0974). Br J Clin Pharmacol 69:15–22PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Vermeersch S, Benschop RJ, Van Hecken A, Monteith D, Wroblewski VJ, Grayzel D, de Hoon J, Collins EC (2015) Translational Pharmacodynamics of Calcitonin Gene-Related Peptide Monoclonal Antibody LY2951742 in a Capsaicin-Induced Dermal Blood Flow Model. J Pharmacol Exp Ther 354:350–357PubMedCrossRef Vermeersch S, Benschop RJ, Van Hecken A, Monteith D, Wroblewski VJ, Grayzel D, de Hoon J, Collins EC (2015) Translational Pharmacodynamics of Calcitonin Gene-Related Peptide Monoclonal Antibody LY2951742 in a Capsaicin-Induced Dermal Blood Flow Model. J Pharmacol Exp Ther 354:350–357PubMedCrossRef
120.
Zurück zum Zitat Rosenfeld MG, Mermod JJ, Amara SG, Swanson LW, Sawchenko PE, Rivier J, Vale WW, Evans RM (1983) Production of a novel neuropeptide encoded by calcitonin gene via tissue-specific RNA processing. Nature 304:129–135PubMedCrossRef Rosenfeld MG, Mermod JJ, Amara SG, Swanson LW, Sawchenko PE, Rivier J, Vale WW, Evans RM (1983) Production of a novel neuropeptide encoded by calcitonin gene via tissue-specific RNA processing. Nature 304:129–135PubMedCrossRef
Metadaten
Titel
Calcitonin gene-related peptide and pain: a systematic review
verfasst von
Wendy Sophie Schou
Sait Ashina
Faisal Mohammad Amin
Peter J. Goadsby
Messoud Ashina
Publikationsdatum
01.12.2017
Verlag
Springer Milan
Erschienen in
The Journal of Headache and Pain / Ausgabe 1/2017
Print ISSN: 1129-2369
Elektronische ISSN: 1129-2377
DOI
https://doi.org/10.1186/s10194-017-0741-2

Weitere Artikel der Ausgabe 1/2017

The Journal of Headache and Pain 1/2017 Zur Ausgabe

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Häufigste Gründe für Brustschmerzen bei Kindern

06.05.2024 Pädiatrische Diagnostik Nachrichten

Akute Brustschmerzen sind ein Alarmsymptom par exellence, schließlich sind manche Auslöser lebensbedrohlich. Auch Kinder klagen oft über Schmerzen in der Brust. Ein Studienteam ist den Ursachen nachgegangen.

Aquatherapie bei Fibromyalgie wirksamer als Trockenübungen

03.05.2024 Fibromyalgiesyndrom Nachrichten

Bewegungs-, Dehnungs- und Entspannungsübungen im Wasser lindern die Beschwerden von Patientinnen mit Fibromyalgie besser als das Üben auf trockenem Land. Das geht aus einer spanisch-brasilianischen Vergleichsstudie hervor.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.