throbber
Review
`
`Molecular studies of CGRP and the
`CGRP family of peptides in the central
`nervous system
`
`Cephalalgia
`2019, Vol. 39(3) 403–419
`! International Headache Society 2018
`Article reuse guidelines:
`sagepub.com/journals-permissions
`DOI: 10.1177/0333102418765787
`journals.sagepub.com/home/cep
`
`Erica R Hendrikse1, Rebekah L Bower1,þ
`Christopher S Walker1
`
`, Debbie L Hay1,2
`
`and
`
`Abstract
`Background: Calcitonin gene-related peptide is an important target for migraine and other painful neurovascular
`conditions. Understanding the normal biological functions of calcitonin gene-related peptide is critical to understand
`the mechanisms of calcitonin gene-related peptide-blocking therapies as well as engineering improvements to these
`medications. Calcitonin gene-related peptide is closely related to other peptides in the calcitonin gene-related peptide
`family of peptides, including amylin. Relatedness in peptide sequence and in receptor biology makes it difficult to tease
`apart the contributions that each peptide and receptor makes to physiological processes and to disorders.
`Summary: The focus of this review is the expression of calcitonin gene-related peptide, related peptides and their
`receptors in the central nervous system. Calcitonin gene-related peptide is expressed throughout the nervous system,
`whereas amylin and adrenomedullin have only limited expression at discrete sites in the brain. The components of two
`receptors that respond to calcitonin gene-related peptide, the calcitonin gene-related peptide receptor (calcitonin
`receptor-like receptor with receptor activity-modifying protein 1) and the AMY1 receptor (calcitonin receptor with
`receptor activity-modifying protein 1), are expressed throughout the nervous system. Understanding expression of the
`peptides and their receptors lays the foundation for more deeply understanding their physiology, pathophysiology and
`therapeutic use.
`
`Keywords
`Amylin, adrenomedullin, calcitonin, CGRP, central nervous system, migraine
`
`Date received: 18 November 2017; revised: 19 February 2018; accepted: 26 February 2018
`
`Introduction
`
`Calcitonin gene-related peptide (CGRP) is an import-
`ant sensory neuropeptide that is involved in pain modu-
`lation (1,2). CGRP has attracted particular interest in
`regard to its role in migraine and likely plays a role in
`other primary headache disorders and painful condi-
`tions (2,3). Presently, three investigational classes of
`drug aim to reduce CGRP activity to prevent and
`treat migraine (3). These are small molecule antagonists
`against a CGRP receptor and antibodies that either
`block receptor activity or bind directly to the CGRP
`peptide. This ‘first generation’ of CGRP-based treat-
`ments will
`likely lead to further drugs over time,
`which will result from a deeper knowledge of the
`CGRP system at a cellular and molecular level and a
`greater understanding of the molecular neuroanatomy
`of CGRP and its receptors. This information guides
`
`understanding of the relative role of central and periph-
`eral processes in which CGRP participates in the patho-
`physiology of migraine, and other disorders.
`Understanding CGRP biology is complex due to the
`existence of similar peptides in the same family, and
`shared receptor complexes. The aim of this review is
`
`1School of Biological Sciences, University of Auckland, Auckland,
`New Zealand
`2Centre for Brain Research, University of Auckland, Auckland,
`New Zealand

`Current address: Department of Pharmacology and Toxicology,
`The University of Otago, Dunedin, New Zealand
`
`Corresponding author:
`Christopher S Walker, School of Biological Sciences, University of
`Auckland, Private Bag 92 019, Auckland 1142, New Zealand.
`Email: cs.walker@auckland.ac.nz
`
`EX2073
`Eli Lilly & Co. v. Teva Pharms. Int'l GMBH
`IPR2018-01427
`
`1
`
`

`

`404
`
`Cephalalgia 39(3)
`
`to summarize information about the expression of
`CGRP and to compare this to its binding sites and
`molecularly defined receptors. We will detail what is
`known about the expression of CGRP and related pep-
`tides, where binding sites for labelled peptides are
`found, and what
`information is available for the
`molecular correlate(s) to that binding. We focus our
`attention on the central nervous system (CNS) and per-
`ipheral ganglia, with emphasis on brain regions that are
`of particular relevance to migraine. The sensory cir-
`cumventricular organs (CVOs) will also be considered,
`as circulating factors can act directly on these sites
`within the brain.
`
`Migraine and the nervous system
`
`Historically, there has been considerable debate around
`the importance of central and peripheral mechanisms of
`migraine. This initially took the form of the competing
`vascular and neuronal hypotheses for migraine. It is
`now clear, based on recent clinical trials, that migraine
`can be treated through peripheral blockade of CGRP
`action (3,4). This does not preclude a central origin of
`migraine. Although the peripheral aspects of the trige-
`minovascular system are important in migraine, it is
`still worthwhile to consider central contributions (5).
`Numerous regions of the CNS are activated during a
`migraine attack and central phenomena are associated
`with migraine symptoms, such as the link between cor-
`tical spreading depression and migraine aura (3). It is
`unclear whether blocking central CGRP action may be
`beneficial or perhaps a hindrance in treating migraine.
`However, central CGRP receptors
`should be an
`important consideration for developing a ‘second gen-
`eration’ of CGRP system-based treatments with poten-
`tially greater effectiveness or fewer side effects.
`This review will focus on the craniofacial pain path-
`way and other brain regions associated with migraine.
`The craniofacial pain pathway begins with Ad and C
`
`fibres of the trigeminal nerve, whose cell bodies are
`located peripherally in the trigeminal ganglia and pro-
`ject centrally primarily into the spinal
`trigeminal
`nucleus (STN) of the brainstem and into the C1/C2
`levels of the spinal cord. The STN is proposed as a
`possible
`site of migraine
`initiation and displays
`increased activity immediately prior to a migraine
`attack (6). Higher order processing of painful signals
`primarily involves the thalamus,
`insular cortex and
`somatosensory cortex.
`Interconnected with these
`regions are other sites, including the amygdala, raphe
`nuclei, periaqueductal gray (PAG), parabrachial area,
`gracile nucleus and locus coeruleus, which play roles in
`pain processing, proprioception, stress or aversion.
`Interestingly, in migraine patients, altered connectivity
`between these regions is reported, such as from the thal-
`amus to the insular cortex and somatosensory cortex or
`from the PAG to the cortex and amygdala (7,8). Other
`brain regions involved in migraine may include the
`hypothalamus,
`implicated in attack initiation;
`the
`hippocampus, which displays greater pain-induced
`activity in migraine patients; and the cortex, cerebellum
`and visual network, which may be involved in cortical
`spreading depression and symptoms of migraine aura
`(9–11).
`
`The calcitonin gene-related peptide family
`of peptides
`
`CGRP belongs to a small family of structurally related
`peptides (Figure 1). There are two forms of CGRP, a
`and b. The broad term ‘‘CGRP’’ is used for either a or
`b, unless specifically noted. The other major members
`of this family are amylin, adrenomedullin (AM) and
`adrenomedullin 2 (AM2). Another member of this
`family,
`the calcitonin receptor-stimulating peptide
`(CRSP) was reported in some mammals (12); however,
`CRSP is not expressed in primates or rodents. Each of
`these peptides contains a conserved pair of cysteine
`
`aCGRP
`bCGRP
`CT
`AM
`AM2
`Amylin
`
`AC-DTATCVTHRLAGLLSRSGGVVKNN-FVPTN-VGSKAF–NH2
`AC-NTATCVTHRLAGLLSRSGGMVKSN-FVPTN-VGSKAF–NH2
`CGNLSTCMLGTYTQDFNKFHTF-------PQTAIGVGAP–NH2
`GC-RFGTCTVQKLAHQIYQFTDKD-KDNVAPRSKISPQGY–NH2
`GC-VLGTCQVQNLSHRLWQLMGPAGRQDSAPVDPSSPHSY–NH2
`KC-NTATCATQRLANFLVHSSNNFGAI-LSSTN-VGSNTY–NH2
`
`Figure 1. Amino acid sequences of calcitonin gene-related peptide family members. Alignment of the amino acid sequences for
`human aCGRP, bCGRP, calcitonin (CT), AM, AM2 and amylin using the single letter code. AM and AM2 have been truncated at the
`N-terminal. All the peptides have a C-terminal amide (-NH2). A conserved disulfide bond between the two N-terminal cysteine
`residues is indicated by a solid line.
`
`2
`
`

`

`Hendrikse et al.
`
`405
`
`form a disulfide bond, creating a
`that
`residues
`loop. They also all contain a C-terminal amide.
`Amylin and CGRP are the most closely related pep-
`tides in terms of amino acid sequence, resulting in over-
`lapping actions
`in pain modulation and nutrient
`balance (13,14). The similarities between the peptides
`within the CGRP family causes significant overlap in
`their ability to activate each other’s receptors. This
`‘‘blurred’’ receptor pharmacology is particularly evi-
`dent at rodent receptors, where there is noticeable
`cross-reactivity. This makes working with this peptide
`family challenging because it is often very difficult to
`determine which of the many possible molecularly
`defined receptors actually mediates an effect for a
`given peptide (15).
`
`Receptor composition and pharmacology
`
`The molecular composition of the CGRP family of
`receptors is illustrated in Figure 2, which highlights
`the overlapping activity of the peptides at the different
`receptors. Overlap occurs, not only because the pep-
`tides have shared features but also because the recep-
`tors
`themselves have shared and closely related
`components. All of the receptors within this family
`are G protein-coupled receptors (GPCRs). Two specific
`GPCRs form high affinity receptors for the different
`peptides due to their association with accessory pro-
`teins. There are three of these accessory proteins, recep-
`tor activity-modifying protein (RAMP) 1, 2 and 3. The
`CGRP, AM1 and AM2 receptors consist of the calci-
`tonin receptor-like receptor
`(CLR) and RAMP1,
`
`CGRP Receptors
`
`Adrenomedullin receptors
`
`CGRP
`
`CGRP > AM
`
`AM1
`AM > AM2
`
`Amylin receptors
`
`AM2
`AM = AM2
`
`AMY1
`CGRP = Amylin
`
`Calcitonin receptor
`
`AMY2
`Amylin
`
`AMY3
`Amylin
`
`Legend
`
`CTR
`
`CT > Amylin
`
`CLR
`
`CTR
`
`RAMP1 RAMP2 RAMP3
`
`Figure 2. The calcitonin gene-related peptide (CGRP) receptor family. The relative potency of CGRP, calcitonin (CT), AM, AM2 or
`amylin is shown below a schematic representation of the appropriate receptor complex (17). The potential overlap between the
`CGRP and amylin receptors is highlighted in purple. The receptor components are described in the legend.
`
`3
`
`

`

`406
`
`Cephalalgia 39(3)
`
`RAMP2 or RAMP3, respectively (16,17). The AMY1,
`AMY2 and AMY3 receptors consist of the calcitonin
`receptor (CTR) with each RAMP (17,18). CLR alone
`does not appear to act as a functional receptor, whereas
`CTR is the receptor for calcitonin (16,17). Figure 2
`shows that CGRP can activate both CLR/RAMP1
`and CTR/RAMP1 equally. CGRP is weaker at other
`human receptor complexes, but there are clear devia-
`tions in other species (15). At rat CLR and CTR-based
`receptors, CGRP is potent at both RAMP3-based
`receptors (19). Amylin can potently activate CTR/
`RAMP1, CTR/RAMP2 and CTR/RAMP3 but not
`the CLR-based receptors. AM activates all
`three
`CLR/RAMP complexes but has a preference for
`CLR/RAMP2 and 3 (17).
`One pharmacological method for teasing apart clo-
`sely-related receptors with overlapping peptide binding
`parameters is to use specific antagonists. Although sev-
`eral antagonists have been reported that are capable of
`antagonizing members of the CGRP receptor family,
`they generally lack the required specificity. For exam-
`ple, although CGRP8-37, an N-terminally truncated
`form of CGRP, is often cited as a specific CGRP recep-
`tor antagonist, it does not display sufficient specificity
`to be practically useful. CGRP8-37 is less than 10-fold
`selective for CGRP receptors over AMY1 and is cap-
`receptors
`able of antagonizing AM2 and AMY3
`(19–22). Similarly, CTR and amylin receptor antagon-
`ists including truncated salmon calcitonin (sCT8-32) and
`AC187 are potent antagonists, but do not distinguish
`between amylin receptor subtypes (19,21). The develop-
`ment of the small molecule ‘‘gepant’’ class of drugs has
`yielded some useful tools for separating the CGRP
`receptor from AM receptors and,
`if used carefully,
`receptors
`can tell apart
`the CGRP and AMY1
`(Table 1). For instance, olcegepant displays greater
`than 10,000-fold selectivity for the CGRP over AM
`receptors and 100–200 fold selectivity for the CGRP
`receptor over the AMY1 receptor (23–25). However,
`other gepants including telcagepant and MK-3207 are
`less selective and the degree of selectivity measured may
`
`depend on other variables, including the signaling path-
`way measured (23,26). With the careful selection of sev-
`eral different agonist and antagonist concentrations, it
`may be possible to pharmacologically characterize
`members of the CGRP receptor family present in a
`particular cell
`line or tissue. However, for practical
`use in in vivo systems more specific pharmacological
`tools are required.
`
`Challenges associated with determining the molecular target for
`a given peptide. To understand the biology of the CGRP
`peptide family, it is crucial to define the molecular iden-
`tity of the peptide and receptor responsible for a bio-
`logical effect in physiological systems. The overlapping
`pharmacology described above represents a major chal-
`lenge associated with studying this important family of
`receptors. Therefore, pharmacological
`approaches
`should be complemented with other methods. The
`measurement of mRNA can be a useful guide.
`However, the detection of mRNA does not necessarily
`mean that protein is present, especially in neurons
`where proteins can be transported to projections dis-
`tant from the cell body (27). Stable membrane proteins
`including CLR, CTR and RAMPs may not need high
`levels of mRNA expression to maintain protein expres-
`sion in steady-state cells. This has been illustrated
`where moderate to high levels of immunoreactivity
`were observed in regions where the corresponding
`mRNA was low or not detected (28,29).
`The development of techniques including high-reso-
`lution confocal
`imaging has allowed researchers to
`detect the precise cellular localization of a receptor
`using antibodies. However, when used to directly
`detect protein expression, antibodies also have limita-
`tions. They require comprehensive validation to con-
`firm specificity and selectivity for their targets (30). In
`many cases, antibodies are used without sufficient evi-
`dence of validation and it may not be possible to draw
`the conclusions that the authors suggest. For example,
`studies may simply lack the required information
`regarding the antibodies used to allow evaluation of
`
`Table 1. Summary of small molecule CGRP receptor antagonists (gepants) for migraine treatment.
`
`Small molecule
`
`Olcegepant
`
`Telcagepant
`
`MK-3207
`
`BHV-5000
`
`Atogepant
`
`Ubrogepant
`
`Rimegepant
`
`Blocks CGRP
`receptor
`Blocks AMY1
`receptor1
`Clinical
`development
`
`Yes
`
`Yes
`
`Yes
`
`Yes
`
`Yes
`
`Yes
`
`Yes
`
`Yes (100–200
`fold lower)
`
`Development
`stopped
`
`Yes (50–100
`fold lower)
`
`Development
`stopped
`
`Yes (50
`fold lower)
`
`Development
`stopped
`
`No data
`
`No data
`
`No data
`
`No data
`
`Preclinical
`
`Phase 2
`
`Phase 3
`
`Phase 3
`
`No data, no published data is available.
`1The relative affinity or potency for the AMY1 receptor compared to the CGRP receptor is reported in parenthesis (20,23,26,154).
`
`4
`
`

`

`Hendrikse et al.
`
`407
`
`the data (31) or the antibodies used have been shown to
`recognize multiple proteins. This prevents observed
`immunoreactivity from being conclusively associated
`with a single protein (30). The latter situation has
`been a significant issue for antibodies designed to
`detect RAMPs (32).
`Even if a receptor subunit has been identified with a
`well-validated antibody, this alone is not meaningful
`for this receptor family, except when considering calci-
`tonin activity at CTR alone. Expression of CLR or
`CTR by themselves says little about
`the receptor
`phenotype without
`further examining potential co-
`localization with a RAMP. RAMPs are also known
`to modify the signaling of other receptors, forming
`alternative complexes that complicate interpretation
`(33). However, valuable information has been obtained
`from studies showing co-localization between CLR and
`receptor-component protein or between CLR and
`RAMP1, indicating the likely presence of the CGRP
`receptor (34–36). The ideal solution maybe to develop
`antibodies that specifically target the heteromeric com-
`plex. This strategy was employed to detect the CLR/
`RAMP1 complex in the trigeminal ganglia, dura mater
`and the spinal trigeminal nucleus (37). However, anti-
`bodies that recognize heteromeric complexes are diffi-
`cult
`to generate and characterize. Thus, protein
`expression data should be used alongside other experi-
`mental observations. For example, mRNA and peptide
`binding correlations were informative in early studies
`validating CLR/RAMP complexes as receptors for
`CGRP and AM (38). Another consideration relates to
`the level of expression reported. For many receptors,
`there is considerable amplification in signal following
`ligand-receptor binding. Hence, relatively low expres-
`sion does not automatically translate
`into little
`function.
`Regardless of method, much research relies on quali-
`tative description of the biological target within ana-
`tomical locations. This is a highly subjective process,
`and is open to individual interpretation of intensity
`and location. Collating data, improving its accessibility,
`and standardisation all help to address these issues. For
`example, the Human Protein Atlas, the Allen Brain
`Atlas and other web-based tools provide excellent
`resources for expression data, assisting identification
`of patterns (39–41).
`
`Peptide accessibility to the CNS and relevance of receptors in
`different locations. The blood-brain barrier (BBB) is an
`important consideration when looking at the origin,
`expression and activity of neuropeptides in the CNS.
`Peptides present in the blood may cross the BBB either
`through active transport or passive diffusion (42,43).
`Alternatively, circulating peptides can interact directly
`with CVOs. CVOs are highly vascularized, with
`
`fenestrated capillaries, allowing peptides access to
`these discrete parts of the CNS. The CGRP family of
`peptides do not freely cross the BBB (44). The propor-
`tion of peripheral amylin that can cross the BBB is low
`and is unlikely to be sufficient to activate receptors
`inside
`the BBB at physiological
`concentrations
`(45,46). Interestingly, the highest amount of labelled
`amylin that was reported to penetrate the brain was
`observed in the hypothalamus and medulla (46),
`which are associated with sensory CVOs and thus are
`permeable to circulating hormones. Hence, relatively
`high levels of amylin experimentally reaching these
`regions is not surprising. The reported actions of
`exogenous amylin at sites inside the BBB, such as the
`ventral tegmental area, are unlikely to be explained by
`amylin crossing the BBB (47–49). Alternative explan-
`ations could include local production of amylin or
`another member of the CGRP peptide family triggering
`receptor activation in these regions. The BBB pene-
`trance of CGRP and AM have been examined primar-
`ily in vascular models, which suggest that neither
`peptide can cross the BBB when the vascular endothe-
`lium is intact (50–52). A more definitive study has been
`performed for AM, which, under normal conditions,
`does not appear to cross the BBB and penetrate the
`brain (53). Curiously, definitive experiments have not
`been performed for CGRP. However, given the lack of
`brain penetrance displayed by calcitonin, amylin and
`AM and data from vascular models, significant cross-
`ing of the BBB by CGRP seems unlikely. Overall, the
`low BBB permeability for CGRP family peptides sug-
`gests that CNS expression of these peptides is required
`for them to have activity inside the BBB.
`
`AM and AM2
`
`AM and AM2 expression in the CNS. AM is best known as
`a regulator of the cardiovascular and lymphatic sys-
`tems. It is a potent vasodilator and is highly expressed
`in the vascular endothelium (54,55). Vascular AM may
`be involved in maintaining the BBB, cerebral circula-
`tion and the volume of cerebrovascular fluid (56–58).
`Expression in the vasculature may complicate expres-
`sion analysis
`that
`relies on homogenized tissue,
`including mRNA analysis and membrane binding.
`mRNA and immunoreactivity for AM has been
`reported in the human and rat brain, localized to the
`vasculature, the choroid plexus and in neurons and glia
`of the hypothalamus, cerebellum and medulla (59–63).
`However, the cerebellum, which has been implicated in
`AM-regulated blood pressure control, was the only
`brain region where mature AM peptide was detected
`(59,64). Interestingly, in a genetic mouse model that
`was modified to lack AM expression in the CNS,
`altered responses to pain, anxiety and stress were
`
`5
`
`

`

`408
`
`Cephalalgia 39(3)
`
`observed (65,66). Similarly, AM has been reported to
`induce pain, and AM-like immunoreactivity was
`detected in dorsal root ganglia neurons and axon ter-
`minals in the dorsal horn from rats (67,68). These stu-
`dies suggest that AM may play a physiological role in
`the CNS despite limited expression.
`The physiology of AM2 is poorly understood; how-
`ever, central or peripheral administration can modulate
`blood pressure (69). AM2 mRNA and immunoreactiv-
`ity has been found in the pituitary and hypothalamus of
`humans and rats as well as the spinal cord and dorsal
`root ganglia of rats (70–74). AM2 may be involved in
`the hypothalamo-pituitary axis and the central control
`of blood pressure in the hypothalamus (69,75–77).
`
`AM and AM2 binding sites in the CNS. AM binding sites
`have been measured in rat and human brain homogen-
`ates, with high levels of 125I-AM binding in the hypo-
`thalamus, thalamus and spinal cord (72,78–80). AM2
`binding sites in the CNS have not been specifically stu-
`died. More work is required to determine the preva-
`lence and location of AM and AM2 binding sites
`within the brain.
`
`Molecular composition of AM and AM2 binding sites in the
`CNS. CLR, RAMP2 and RAMP3 are the most relevant
`to the AM peptides (Figure 2; Hay et al., 2017). The
`expression of CLR mRNA and protein has been stu-
`died extensively within the nervous system, where it is
`widespread (Table 2 (28,70,81–83)). Thus, the presence
`of functional AM1 and AM2 receptors is dependent
`upon the expression of RAMP2 or RAMP3, respect-
`ively. The expression of mRNA for these receptor com-
`ponents has been well documented in the vasculature
`(50,84,85). However, there is only limited data describ-
`ing RAMP2 or RAMP3 in discrete rat nervous system
`regions (Table 2). In rats, abundant mRNA for
`RAMP2 was detected in the hypothalamus, and
`RAMP3 was detected in nuclei of
`the thalamus
`(86,87). Relatively high RAMP2 expression was also
`detected in the hippocampus and spinal cord (70,86).
`RAMP2 and RAMP3 have not been examined in a
`human brain (Table 3). Unfortunately, there are cur-
`rently no antibodies for the RAMP2 or RAMP3 pro-
`teins that have been sufficiently characterized for use in
`immunohistochemical mapping (33). Characterized
`antibodies that have been used for the detection and
`mapping of CLR, CTR and RAMP1 are discussed
`later.
`
`Calcitonin
`
`Calcitonin expression in the CNS. Canonically, calcitonin is
`expressed by thyroid C cells and regulates calcium
`(88–90). The CALCA gene
`homeostasis
`encodes
`
`calcitonin and CGRP mRNA (91). Tissue-specific alter-
`native splicing of CALCA mRNA results in the prefer-
`ential expression of CGRP mRNA in the nervous
`system; calcitonin mRNA is not detectable in these
`regions (1,88,92,93).
`
`Calcitonin binding sites in the CNS. Although calcitonin
`does not appear to be expressed in the nervous
`system, binding sites for calcitonin are found in many
`brain structures (94–96). Salmon calcitonin has fre-
`quently been used as a tool in calcitonin studies because
`it has higher affinity than human or rat calcitonin and
`can bind CTR alone, AMY1, AMY2 and AMY3 recep-
`tors with high affinity (18). Thus, depending upon the
`presence or lack of a RAMP, salmon calcitonin binding
`may indicate the presence of a receptor for amylin,
`CGRP or calcitonin. Salmon calcitonin binding sites
`have been examined in rat and primate nervous systems
`using autoradiography (Tables 2 and 3). The data are
`generally consistent between these studies with only
`minor differences. Overall, autoradiography shows
`high levels of salmon calcitonin binding in the hypo-
`thalamus, sensory CVOs and the nucleus accumbens,
`along with moderate binding in the bed nucleus of the
`stria terminalis (BNST), PAG and locus coeruelus
`(95,97–100). The spinal cord has minimal salmon calci-
`tonin binding in the dorsal horn (95,97). There also
`seems to be very little binding in the cerebral cortex
`and in the cerebellum. Data from the human brain is
`more limited. In a single study, salmon calcitonin bind-
`ing was examined by autoradiography in the human
`medulla (Table 3). Discrete binding sites were observed
`in several nuclei, including high densities of binding in
`the NTS and raphe nuclei. Very little binding was
`observed in the fibre tracts (96). In another study,
`human hypothalamic membranes showed high levels
`of salmon calcitonin binding (101). The variability in
`displacement of salmon calcitonin by human calcitonin
`between brain regions suggests that AMY receptors
`account for a significant proportion of salmon calci-
`tonin binding sites (102,103). However, more work is
`required to determine the prevalence and location of
`calcitonin binding sites within the human brain, and
`their direct relevance to calcitonin, amylin and/or
`CGRP activity.
`
`in the
`Molecular composition of calcitonin binding sites
`CNS. Consistent with widespread calcitonin binding
`sites, CTR mRNA and protein expression has been
`observed throughout the nervous system of rodents
`(Table 2). Several splice variants of the CTR have
`been reported. These variants are not necessarily con-
`served across species and can display altered functions
`(104). This review does not distinguish between these,
`because there are no tools to determine the expression
`
`6
`
`

`

`Hendrikse et al.
`
`409
`
`(81,86,87,97,98,115,145,146,150,151,158,170)
`
`(29,81,86,87,97,99,105,115,139,145,162,170)
`
`(29,81,86,87,115,139,145,147,151,158,170)
`
`(29,86,87,92,97-99,105,115,139,145,146,148,
`
`151,152,162-164,170,171)
`
`(86,87,92,97-99,105,115,139,145,146,148,151,
`
`(81,86,87,92,99,115,145,146,150,151,162,169)
`
`152,162-164,170)
`
`148,150,158,162-164,168)
`
`(29,81,86,87,92,97,98,105,114,115,139,145-
`
`MP
`
`(29,86,92,97,98,105,115,139,145,153,162)
`
`164,167)
`
`(29,47,81,92,97-99,105,115,139,145-148,162-
`
`(29,81,87,92,97,105,115,145-147,162,163,166)
`
`(81,86,87,97-99,105,115,143,145-148,151)
`
`145-148,150,151,158,162-164)
`
`(29,81,82,86,87,92,97-99,105,115,132,139,
`
`146,158,161,165)
`
`(29,67,70,82,86,87,92,97,101,131,139,140,
`
`P
`
`(115,151)
`
`(115)
`
`(87,99,115,139,164)
`
`(99,115,139,145,146,162)
`
`(87,99,115,145,146,150,151)
`
`(99,105,115,139,145,146,150,151,162,163)
`
`(29,45,70,107,128,131,158-161)
`
`(20,29,35,92,93,132,133,138,139,155-158)
`
`MP
`
`P
`
`P
`
`P
`
`MP
`
`MP
`
`P
`
`MP
`
`MP
`
`P
`
`MP
`
`MP
`
`P
`
`MP
`
`MP
`
`P
`
`P
`
`P1
`P1
`
`MP
`
`MP
`
`References
`
`Peptide
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`N
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`N
`
`Y
`
`N
`
`N
`
`N
`
`Y
`
`Y1
`
`Y
`
`N
`
`Y
`
`Y
`
`N
`
`Y
`
`Y
`
`Y
`
`Y
`
`N
`
`N
`
`N
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`M
`
`125I-sCTCGRPAmylin
`
`125I-Amy
`
`125I-CGRP
`
`Bindingsites
`
`1Verylowexpression.
`ordatanotavailable;CVOs:circumventricularorgans;OVLT:organumvasculosumoflaminaterminalis.
`M:mRNAdetectedbypolymerasechainreactionorinsituhybridisation;P:protein/immunoreactivityintissues;Y:specificradioligandbindingdetected;N:nobindingdetected;Emptyspace:noexpression
`
`M
`
`MP
`
`MP1
`
`P
`
`M
`
`M
`
`P
`
`P
`
`Cerebralcortex
`
`Septalarea
`
`Hippocampus
`
`Basalganglia
`
`Amygdala
`
`Thalamus
`
`MP
`
`MP
`
`Hypothalamus
`
`MP
`
`MP
`
`MP
`
`P
`
`grey
`
`P
`
`Periaqueductal
`
`P
`
`MP
`
`Midbrain
`
`Pons
`
`Cerebellum
`
`MP
`
`MP
`
`Medulla
`
`MP
`
`MP
`
`Spinalcord
`
`Pinealgland
`
`organ
`
`Subcommisural
`
`Medianeminence
`
`Pituitary
`
`OVLT
`
`Subfornicalorgan
`
`Areapostrema
`
`CVOs
`
`P
`
`MP
`
`MP
`
`Dorsalrootganglia
`
`Trigeminalganglia
`
`CLRCTR
`
`Region
`
`Receptorsubunit
`
`Table2.ExpressionofCGRPfamilyreceptorcomponentsandpeptidesandtheirbindingsitesinrodents.
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`M
`
`MP
`
`P
`
`MP
`
`M
`
`M
`
`MP
`MP1
`
`M1P
`
`M
`
`M1
`
`M
`
`MP
`
`P
`
`MP
`
`MP
`
`MP
`
`M
`
`M
`
`M
`
`MP1
`MP
`
`M
`
`M
`
`M
`
`MP
`
`P
`
`RAMP3
`
`RAMP2
`
`RAMP1
`
`7
`
`

`

`410
`
`Cephalalgia 39(3)
`
`Table 3. Expression of CGRP family receptor components and peptides and their binding sites in primates including humans.
`
`Receptor subunit
`
`Binding sites
`
`Peptide
`
`Region
`
`CLR CTR RAMP1 RAMP2 RAMP3
`
`125I-CGRP
`
`125I-Amy
`
`125I-sCT CGRP Amylin References
`
`Trigeminal ganglia
`
`MP
`
`P
`
`P
`
`Dorsal root ganglia MP
`
`CVOs
`
`Area postrema
`
`MP
`
`P
`
`MP
`
`Subfornical organ
`
`OVLT
`
`Pituitary
`
`MP
`
`Median eminence MP
`
`Subcommisural
`organ
`
`Pineal gland
`
`Spinal cord
`
`Medulla
`
`MP
`
`MP
`
`MP
`
`P
`
`MP
`
`MP
`
`MP
`
`MP
`
`MP
`
`Y
`
`Y
`
`Y
`
`N
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`N
`
`MP
`
`MP
`
`(20, 35, 37, 83, 133, 172)
`
`(140)
`
`(28, 95, 96, 106, 163)
`
`(95)
`
`(95)
`
`(28, 101)
`
`(28)
`
`(28)
`
`P
`
`P
`
`P
`
`(28, 82, 95, 101, 140)
`
`(20, 28, 37, 82, 95, 96,
`106, 147, 163, 173)
`
`(34, 95, 101, 147)
`
`MP
`
`MP
`
`MP
`
`MP
`
`MP
`
`Cerebellum
`
`Pons
`
`Midbrain
`
`MP
`
`MP
`
`MP
`
`Periaqueductal grey MP
`
`MP
`
`P
`
`Hypothalamus
`
`Thalamus
`
`Amygdala
`
`Basal ganglia
`
`Hippocampus
`
`Septal area
`
`Cerebral cortex
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`P
`
`P
`
`NS
`
`Y
`
`Y
`
`N
`
`Y
`
`Y
`
`Y
`
`Y
`
`NS
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`Y
`
`(28, 95, 147, 163, 173)
`
`(28, 95, 101, 147, 163)
`
`(28, 95, 147, 173)
`
`(28, 95, 101, 147, 163)
`
`(95, 147)
`
`(95, 147, 163)
`
`(95, 147, 163)
`
`(95)
`
`(95)
`
`(95)
`
`M: mRNA detected by polymerase chain reaction or in situ hybridisation; P: protein/immunoreactivity in tissues; Y: specific radioligand binding detected;
`N: no binding detected; Blank space: no expression or data not available; NS: non-specific binding; CVOs: circumventricular organs; OVLT: organum
`vasculosum of lamina terminalis.
`1Very low expression or binding.
`
`profile of specific splice variants at the protein level. We
`discuss CTR only in general due to limited data, even at
`the mRNA level. In the peripheral nervous system,
`CTR staining was observed in neuronal cell bodies in
`the
`trigeminal and dorsal
`root ganglia (20,29).
`Centrally, dense protein expression was reported in
`fibres and cell bodies of brain regions including the
`nucleus accumbens, the substantia nigra, the NTS and
`the area postrema (105). Moderate staining of cell
`bodies and fibres was observed in the spinal cord,
`amygdala, PAG and dorsal
`raphe nucleus
`(105).
`Limited but discrete staining has also been observed
`in the thalamus, BNST and regions of the somatosen-
`sory cortex (29,105). Interestingly, CTR protein has
`also been reported in several regions involved in the
`perception and higher-order processing of craniofacial
`pain (Table 2). The pattern of CTR expression in
`human neural tissues is similar to that of rodents,
`although data are limited to the trigeminal ganglia
`and the medulla (Table 3;
`(20,106). Widespread
`
`expression of CTR was observed in the human medulla,
`including the NTS, STN, cuneate, gracile and hypo-
`glossal nuclei (106). Considering the lack of calcitonin
`expression in central and peripheral nervous tissues and
`its limited ability to cross the BBB, it is surprising that
`CTR is so widely expressed within the nervous system.
`This suggests that the CTR probably functions in con-
`cert with one or more RAMPs inside the nervous
`system to bind amylin, CGRP, or another peptide.
`
`Amylin
`
`Amylin expression in the CNS. The pancreatic b-cell is a
`major site of amylin expression and releases amylin into
`the circulation to induce meal-ending satiation (13).
`Early reports show a lack of amylin expression in the
`nervous
`system (107,108). However,
`later work
`reported limited amylin expression within the brain
`and in sensory neurons (Table 2; (109–111). It is
`likely that some amylin antibodies can also detect
`
`8
`
`

`

`Hendrikse et al.
`
`411
`
`CGRP and some CGRP antibodies can detect amylin.
`Potential cross-reactivity between amylin and CGRP
`antibodies may account for some discrepancies between
`studies (112). Hence, care should be taken to confirm
`that antibodies detect only the desired target. More
`recently, amylin mRNA expression was reported in
`the preoptic area and BNST neurons of lactating
`dams (113). Amylin mRNA and immunoreactivity
`has also been reported in hypothalamic neurons in
`mice (114). Although amylin expression has not been
`examined in the human brain (Table 3), the limited
`amylin expression in the CNS suggests that its central
`actions likely occur predominantly outside the BBB
`through the CVOs, although discrete expression in a
`limited number of CNS regions is possible.
`
`Amylin binding sites in the CNS. Despite the reportedly low
`expression of amylin in the CNS, 125I-amylin binds with
`widespread distribution in rat brain wh

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket