AU2001248972A1 - Use of npy y1 receptor antagonists in the treatment of inflammatory conditions - Google Patents

Use of npy y1 receptor antagonists in the treatment of inflammatory conditions

Info

Publication number
AU2001248972A1
AU2001248972A1 AU2001248972A AU4897201A AU2001248972A1 AU 2001248972 A1 AU2001248972 A1 AU 2001248972A1 AU 2001248972 A AU2001248972 A AU 2001248972A AU 4897201 A AU4897201 A AU 4897201A AU 2001248972 A1 AU2001248972 A1 AU 2001248972A1
Authority
AU
Australia
Prior art keywords
mice
npy
receptor
inflammation
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2001248972A
Inventor
Guilherme De Araujo Lucas
Patrik Ernfors
Hassameh Hassani
Philippe Naveilhan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GLOBAL GENOMICS AB
Original Assignee
Karolinska Innovations AB
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Karolinska Innovations AB filed Critical Karolinska Innovations AB
Publication of AU2001248972A1 publication Critical patent/AU2001248972A1/en
Assigned to GLOBAL GENOMICS AB reassignment GLOBAL GENOMICS AB Request for Assignment Assignors: KAROLINSKA INNOVATIONS AB
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2271Neuropeptide Y
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/155Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1796Receptors; Cell surface antigens; Cell surface determinants for hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Endocrinology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

Use of NPY Yl receptor antagonists^in the treatment of inflammatory conditions
Field of the invention
The present invention relates to selective neuropeptide Y (NPY) Yl receptor antagonists. More closely, it relates to use of, and methods of using, NPY Yl receptor antagonists for the treatment of inflammatory conditions.
Background of the invention
Neuropeptide Y (NPY) has a wide range of physiological functions, particularly affecting the cardiovascular system. NPY is also believed to exert anti- nociceptive actions by inhibiting the release of substance P(SP) and other "pain neurotransmitters" in the dorsal horn of the spinal cord 1-2-3. However, the physiological significance and potential therapeutic value remains obscure 4.
NPY is known to bind with high specificity to several receptor subclasses which have different biological functions. Several pharmacological applications of compounds having NPY receptor agonistic or antagonistic effect have been described.
For example, US 6 017 879 describes template-associated NPY Y2-receptor agonists for treatment of asthma, rhinitis, and bronchitis. An other example is US 5 939 462 describing NPY Y5-receptor antagonist for treatment of obesity.
There is no prior art describing selective NPY receptor agonists having anti- nociciptive action and no prior art describing selective NPY receptor antagonists having anti-inflammatory action. To have selective drugs is highly desirable in view of less side effects etc.. Summary of the invention
The present invention provides new therapeutic approaches concerning treatment of inflammatory conditions.
In a first aspect, the invention relates to use of, or method of using, a neuropeptide Y Y 1 receptor antagonist for preparation of a drug for preventing and/or treating inflammatory conditions.
The NPY Yl receptor antagonist may be topically, subcutaneously or systemically administered for the treatment of cutaneous inflammation. In an other embodiment, the NPY Y 1 receptor antagonist is topically or systemically administered for the treatment of internal inflammation. A preferred use is for treatment of neurogenic inflammation. Another use is for treatment of acute or chronic/ persistent inflammation.
When the drug is used systemically in any aspect of the invention, the administration may preferably be orally.
Already available as well as yet unknown compounds having NPY Y 1 antagonistic or inhibiting effect can be used for the purposes of the invention.
Therefore, in a second aspect, the invention relates to use of the NPY Yl receptor as a drug target in screening procedures to find antagonists of said receptor, more precisely to find anti-inflammatory compounds which directly or indirectly affect the NPY Y 1 receptor in a selective way for treatment of the above described inflammatory conditions.
Preferably, high throughput screenings procedures are used to find small organic biocompatible molecules. To identify a possible physiological role for NPY in pain transduction and to identify the particular receptor subtypes involved, the present inventors generated NPY Yl receptor null mutant mice (Yl-/-) by homologous recombination techniques.
The present inventors show that Yl-/- mice develop hyperalgesia to acute thermal, cutaneous and visceral chemical pain and exhibit mechanical hypersensitivity. Neuropathic pain is augmented and the mice show a complete absence of the pharmacological analgesic effects of NPY. In the periphery, Y l receptor activation is sufficient and required for SP release and the subsequent development of neurogenic inflammation and plasma leakage.
The present inventors conclude that the Y 1 receptor is required for central physiological and pharmacological NPY-induced analgesia and that its activation is both sufficient and required for the release of SP and initiation of neurogenic inflammation.
Detailed description of the invention
Homologous recombination in embryonic stem cells was used to establish mice deficient in the NPY Yl receptor. The disruption was generated by introducing an internal ribosomal entry site followed by a Tau-LacZ fusion minigene into the second exon of Yl (Fig. l a). Southern blot analysis confirmed that the Yl allele was disrupted and Northern blot analysis showed that instead of the mRNA transcripts encoding Yl , the mutant (Yl " ") mice produced the expected mRNA encoding β-galactosidase (Fig. lb-d). As previously described, female Y\~'~ mice display a late-onset overweight compared to their littermates (data not shown). Yl receptors are abundant in the forebrain while little or nothing is present in the brainstem . Yl receptors are also highly expressed in dorsal root ganglion neurons in preferentially small and medium size neurons ' . However, the central teπnination of Yl nerve fibers in the dorsal horn, and whether Yl is expressed in both of the two major cytochemical subpopulations of pain neurons, the SP peptidergic and non-peptidergic pain neurons , is unresolved, β-galactosidase histochemical and immunohistochemical staining of spinal cord sections from Y r _ mice led to strong staining localised exclusively to the dorsal horn (Fig. l e and f). Immunohistochemical double staining for β-galactosidase (staining Yl expressing neurons and fibers) and the lectin EB4 (staining somas and nerve fibers of unmyelinated non-peptidergic sensory nociception neurons ) showed a strong staining for Yl nerve fibers in dorsal horn layer II overlapping with IB4 terminals. A significant portion of the nerve fibers also terminated in layers I, III and IV (Fig. If). Yl -positive dorsal horn interneurons were also found (Fig. If, arrows). Many Yl expressing dorsal root ganglion neurons coexpressed SP (Fig. lh; 38% of Yl neurons contained SP), however, a large number of Yl neurons also double stained for IB4 (Fig. lg; 32%).
A presynaptic block of primary afferent SP release in the spinal cord may participate in central NPY-induced analgesia . We therefore examined if this primary afferent circuit was intact in the Yl "7" mice. No overt alteration of SP nor NPY immunoreactivity was detected in the spinal cord of Yl "7" mice (data not shown). Furthermore, total SP in spinal cord measured by El A was 2852±328.9 pg/g tissue and 3919±444.1 pg/g tissue in wild-type and Y l 7" mice, respectively (P>0.05, student's t-test). Immunohistochemical detection of the SP receptor revealed similar staining in the somatic and dendritic cell surface of neurons in the superficial spinal cord lamina (I and II) of both wild- type and Yl" /" mice, as in previous results 9 (Fig. li and j). SP release leads to SP receptor activation and internalisation . Neurons containing internalised SP receptors were detected in Yl7" mice following an intraplantar injection of capsaicin (Fig. Ik and 1), indicating that there is no defect of receptor activation per se in the absence of the Yl receptor. Thus, we conclude that the major neuronal pain circuit suggested to be modulated by NPY is anatomically intact in the Yl7" mice.
Behavioural acute nociceptive thresholds were markedly affected by an absence of Yl . SP/neurokinin A null mutant mice show a blunted response to painful stimulus only at moderate intensities, whereas response to mildly or intensely painful stimulus is intact . We therefore tested whether also the NPY Yl receptor act at specific thresholds in modulating nociception. Withdrawal latency in the hot plate assay was examined at 48, 50, 52, 55 and 58 °C. At 48°C no significant difference was observed (data not shown). The Yl7" mice showed, however, a profound hyperalgesia and displayed a significantly reduced latency at all temperatures above 48°C (Fig 2a). The hot plate test involves supraspinal integration associated with the paw withdrawal. To test whether neuropeptide Yl receptor could be acting in a spinal circuitry, we characterised these mice in the tail flick assay. Consistent with the hot plate assay, Yl7' mice showed a markedly reduced latency at all temperatures between 46-54°C (Fig. 2b). Yl7" mice also displayed a significant decrease in mechanical threshold indicating mechanical hypersensitivity (Fig. 2c).
We then examined a role for the NPY Yl receptor in chemical nociception. During the first phase of the fomialin assay, which provides a measure of the acute pain mediated by direct chemical activation of C-fibers, the nociceptive behaviour was augmented by 44%, 60% and 46% at 1.2, 2 and 5% formalin injected, respectively, in Yl7" mice (Fig. 2d). The second phase of nociception was not consistently altered by an absence of the NPY Y l receptor. In two models of visceral pain, one that is secondary to an inflammatory response (acetic acid) and one that induces immediate pain independent of inflammation (MgSC^) , we also found significantly increased pain behaviour in Yl7" mice (Fig. 2e and f). Combined, the above results indicate that NPY could be acting on Yl containing polymodal nociceptive afferents (C-fibers). Consistent with our results, these neurons span different modalities, and mediate pain transduction from both visceral and cutaneous tissues.
Stress induces analgesia through endogenous opioid and non-opioid dependent mechanisms 12. We tested whether these pathways interact with NPY-dependent analgesia by letting the mice swim in 10°C water producing a non-opioid, NMDA-dependent analgesia and 33°C water resulting in opioid-dependent analgesia ' . After a swim at 10 or 33°C, the development of analgesia assayed in the hot plate assay was similar between wild-type and Yl 7" mice (Fig. 2g). These data suggest that the Yl receptor is not a critical component in stress-induced analgesia. The role of NPY in neuropathic pain is incompletely defined and a number of conflicting results with regards to possible NPY receptors involved have been reported ' . We tested the physiological role of the Yl receptor in a model of neuropathic pain. A partial sciatic nerve ligation resulted in mechanical allodynia (sensitisation to mechanical stimuli) in wild-type mice (37% and 52% increase in sensitivity at 3 and 14 days after nerve injury, respectively, compared to day 0; Fig. 2h). As indicated before, the basal threshold of mechanical sensitivity was significantly decreased in non- lesioned Yl7" mice. Despite this, the mechanical allodynia caused by nerve damage was significantly increased in Yl 7" mice compared to wild-type mice (55% and 67% increase in sensitivity at 3 and 14 days after nerve injury, respectively, compared to day 0; P<0.01 for the slopes of the curves between day 0-14 after nerve injury).
Pharmacological NPY-induced analgesia to thermal stimuli following spinal delivery is well documented ' . To identify the receptor involved in the pharmacological effects of intrathecally administered NPY we injected NPY (10 μg) in the spinal cord of Yl 7" mice and measured heat sensitivity. The anti-nociceptive effect of NPY on the spinal cord was completely abolished in Yl7" mice (Fig. 2i). Thus, the Yl receptor is exclusively responsible for the analgesic effects of centrally delivered NPY.
Inflammation is caused by a neurogenic as well as a non-neurogenic
17 component . Neurogenic inflammation does not occur in the denervated
1 7 i o human skin, and can be prevented by a nerve block in rats ' and is mediated by a peripheral release of SP/ neurokinin A . We tested whether the Yl receptor could participate in inflammation. A subcutaneous injection of capsaicin, which induces neurogenic inflammation, led to a marked inflammation seen by an increased paw diameter, plasma extravasation and hyperalgesia in wild-type mice. Unexpectedly, Yl7" mice displayed no overt or quantitative sign of plasma extravasation, increase in paw diameter and hyperalgesia (Fig. 3a, b, c and d). Neither rectal and paw skin temperature nor heart rate differed between any of the groups before and after capsaicin administration (data not shown). Both wild-type and Yl 7" mice exhibited an increased blood flow in the paw as a response to capsaicin (181,4±31 ,3% and 245,5 ±71 ,4%, respectively) indicating that the Yl receptor is not influencing capsaicin-induced vasodilation. In contrast to capsaicin, mustard oil induces inflammation that is largely, but not exclusively, dependent on a neurogenic component including the release and proinflammatory effects of SP Yl 7" mice showed markedly reduced plasma extravasation in response to mustard oil compared to wild-type mice. However, since the increase in plasma extravasation was significant (although at a lower level), some components of the effects of mustard oil, likely those of non-neurogenic origin, were intact in Yl7" mice (Fig. 3h). In contrast to neurogenic inflammation, there was a similar increase of plasma extravasation, paw diameter and sensitisation following non-neurogenic inflammation induced by carrageenan in Yl7" mice as in wild-type mice (Fig. 3e, f and g).
Since capsaicin and to a large extend mustard oil-induced inflammation depend on the integrity of primary C-fiber afferent release of SP , we deteπnined if Yl is required prior or after SP release in the sequence of events leading to inflammation by injecting SP in the paw. SP caused a similar inflammatory response in Yl 7" mice as it did in wild-type mice (Fig.
3i).
SP receptor immunoreactivity was intact in the skin of wild-type and
Y l " mice and was found occasionally in nerve endings in deπnis and in scattered cells throughout deπnis coπesponding to mast cells, similar to wild- type mice (data not shown). Our results are therefore consistent with that Yl could be required for capsaicin induced SP release. We measured by EIA the quantity of total and released SP in the skin after capsaicin injection in wild- type and Yl7" mice. Capsaicin caused a marked increase of released SP in the skin of wild-type mice whereas it had no effect on SP release in Y 1 ~'~ mice (Fig. 4a). The lack of an increase of released SP was not caused by an overall reduction of SP peripherally because the quantity of total SP was similar in Yl7" and wild-type mice (2269±524 pg/g tissue and 2520±860 pg/g tissue, respectively). Furtheπnore, the number of SP immunoreactive nerve fibers in the deπnis and epidennis was similar in wild-type and Yl 7" mice (5.7±0.5 and 6.2±0.5 per cm, respectively). In accordance with previous results, capsaicin led to a marked reduction of immunoreactive terminals in the epidennis of wild-type mice (from 3.0±0.3 to 1.8±0.2 per cm; P<0.01, student's t-test) possibly by a loss of immunoreactivity due to increased release. In contrast, Yl7" mice displayed no reduction in SP immunoreactive terminals (3.1±0.3 and 2.5±0.6 per cm, respectively). These results indicate that the absence of neurogenic inflammation in Yl7" mice is caused by the requirement of Yl activation for SP release. The persistent vasodilation in these mice could be caused by a normal release of calcitonin gene related peptide, which induces vasodilation but not extravasation. Furthermore, since close to one order of magnitude less SP is required for vasodilation than for plasma leakage21, a small residual SP release in these mice could also cause this phenotype.
We challenged our results in wild-type mice by injecting the Yl agonist [Leu31-Pro 4]-NPY and by using a Yl receptor antagonist. [Leu31- ProM]-NPY efficiently caused plasma extravasation in wild-type mice, and consistent with its specificity for the Yl receptor, no response was seen in Yl 7" mice (Fig. 4b). Administration of the Y l antagonist BIBP 3226 prior to intraplantar injection of capsaicin markedly reduced plasma extravasation in wild-type mice (Fig. 4c), showing that the results on the Yl 7" mice are directly caused by the absence of Yl signalling. We therefore conclude that the NPY Yl receptor is both required and sufficient to induce neurogenic inflammation by controlling SP release, and that a Yl antagonist could provide an effective strategy for the treatment of neurogenic inflammatory diseases.
We have shown that the Yl receptor play an essential anti-nociceptive role during pain transduction in many modalities including thermal, chemical and mechanical from both cutaneous and visceral tissues as well as during neuropathic pain. NPY or another Yl receptor ligand22 could mediate antinociception by reducing SP and excitatory neurotransmitter release from primary C-fiber afferents 3 ' 23 ' 24 and/or by inhibiting post synaptically the SP receptor expressing projection neurons of the spinal cord ' . Consistent with that NPY does not modulate pain transmission only through a presynaptic regulation of SP release, the nociceptive phenotype of the Yl7" mice does not fully correlate with SP and SP receptor null mutant mice. SP receptor null
77 mutant mice show for instance a reduced stress-induced analgesia . We also show that Y l receptor activation is both sufficient and required for neurogenic inflammation. Because mustard oil-induced inflammation occur independent of the vanilloid receptor that is activated by capsaicin19, our results suggest that activation of the Yl receptor could be a shared and obligatory component in most, or all, neurogenic inflammatory conditions.
Methods
Y l gene targeting. Exon 2 of the Yl gene was partially deleted and replaced by a IRES-tau-lacZ cassette also containing a neomycin-resistance gene driven by the PGK promoter and poly A (ETLN). A 0.7 kb DNA fragment 3' from the Yl targeting construct was used as external probe. Homologous recombinant embryonic stem cells clones were injected to generate Yl mutant 129SVXBalb/c hybrid mice. Mice were analysed by Southern blot and PCR using the pπmers 5'-ATCAAATTCTGACCGACGAG-3\ 5'- CATGATGTTGATTCGCTTGG-3 and 5 '-
GCAGCCTCTGTTCCACATACA-3'. Standard procedures were used for Northern blot analysis and 60 μg/sample of total RNA from adult brain was analysed.
Physiological studies. Heart rate and body temperature was measured as
28 previously described . Skin blood flow and temperature was monitored with a thermal probe (Physitemp bat-12) subcutaneously inserted in the plantar region of the paw and a Laser doppler flowmeter probe applied to the plantar surface (Permided PF2B) was used to measure basal and capsaicin-evoked changes. Student t-test was used and differences were considered significant at P< 0.05. Behavioural studies. Between 6-13 adult male F2-3 129SVxBalb/c mice of each genotype were used for all studies. Thermal", mechanical", chemical", visceral" sensitivity, stress-induced analgesia27 and neuropathic pain "1 was assessed as previously. Tail-flick latency (by directing a concentrated light beam to the tail of the mouse) was monitored before and after intrathecal injection of 10 μg NPY. For evans blue plasma extravasation the following were used: capsaicin, 3 μg in 10 μl (Sigma; dissolved in 5% ethanol, 5% Tween-80 and 90% saline), 1% caπageenan (Sigma; dissolved in saline), SP, 50 pmol/paw (Sigma; dissolved in saline), 5% mustard oil (Fluka; dissolved in mineral oil), NPY Yl receptor agonist [Leu31-Pro34]-NPY, 10 μg/paw (Calbiochem; dissolved in saline and 5% acetic acid) and NPY Yl receptor selective antagonist BIBP 3226, 10 mg/kg in 10 ml/kg (American Peptide; dissolved in saline and 5% acetic acid). Briefly, mice were anaesthetised and injected intravenously with Evan's Blue (50 mg/kg) into the jugular vein. Agents mentioned above were injected into one paw of the animal except for Yl receptor selective anatagonist, BIBP 3226, which was injected intravenously 10 min prior to injection into the paw. The other paw was injected with vehicle. After 30 min the plantar skin of the paw was removed, dried off excess liquid, weighed and incubated in formamide for 24h at 56 °C. Extravasated evans blue was measured by spectrophotometer at 620 nm. Mechanical sensitivity was determined before, 30 min and 3h after capsaicin and carrageenan administration, respectively. Carrageenan inflammation was induced similarly but extravasation was measured after 4 hours. The paw diameter was measured before and after capsaicin, carrageenan or vehicle administration using a spring-loaded calliper. Immunohistochemistry. Wild-type and Y1" mice were perfused with 4% paraformaldehyde (for SP receptor immunohistochemistry, mice were perfused with 4% paraformaldehyde and 12.5% picric acid 10 min after capsaicin injection into hindpaw) and the spinal cord and dorsal root ganglia were sectioned coronally ( 15 μm in thickness). Capsaicin was injected intradeπnally into dorsal skin of mice. After 10 min the skin was removed, postfixed and sectioned as above. Immunohistochemistry was perfonned as previously28 using α-β-galactosidase (1 :200 dilution, ICN/Cappel) rabbit α-
SP ( 1 :5000 dilution, Chemicon), guinea pig -SP (1 :200 dilution, Peninsula
Lab.), rhodamine-conjugated bandeiraea simplicifolia lectin I (Isolectin B4;
1 : 100 dilution, Vector), α-NPY (1 :200 dilution, Peninsula Lab.), and rhodamine or FITC-conjugated secondary antisera (Jackson). For SP receptor immunohistochemistry sections were incubated 30 min in PBS, 50% methanol and 0.6% H 02 prior incubation in 10% goat seπim. The antiserum
(Chemicon 1 :2000) was used in the fluorescein TSA fluorescence system
(NEN). β-galactosidase histochemical staining was perfonned as previously
28
EIA. Capsaicin or saline was injected into the paw of WT or Yl7" mice. After 10 min, the paw was removed and the skin was cut open and washed in PBS and 0.1% BSA for 10 min. The skin was then dried, weighed, transferred to a new container and frozen. The liquid was centrifuged at 4000 rpm for 15 min. Supernatant was transferred to a new tube, weighed and frozen. The lumbar part of spinal cord was removed, weighed and frozen. The samples were then assayed for SP according to the manufacturer's instructions using SP high sensitivity EIA kit (Peninsula Lab.). Figure legends
Figure 1. Targeted mutagenesis of the Yl receptor and expression analysis of Yl and SP receptors, a, Yl gene-targeting. Top, targeting vector (Yl coding exons=black boxes). The dismpting cassette is indicated. Bottom, restriction map of the resulting targeted allele (B-BamHI; Sp-Spel; E-EcoRI; P-Pacl; Pr, probe used in the Southern blots), b, Southern blot analysis of ES cells, c, PCR genotyping of wild-type, Yl+/" and Yl " mice. d, Northern blot analysis of total brain RNA of Yl+/+ and Yl7" mice using a Yl probe (Yl Pr) or LacZ probe (LacZ Pr). Probes used are underlined in red in (a), e, A transverse section from the spinal cord lumbar enlargement of Yl7" mice histochemically stained for β-galactosidase. f, Immunohistochemical staining of Yl7" mice for β-galactosidase-positive nerve terminals and neurons (arrows) in the spinal cord dorsal horn (green) and the lectin IB4 (red, layer Ilinner). g, Double staining of L4 dorsal root ganglion for β-galactosidase (green) and IB4 (red), h, Double staining of L4 dorsal root ganglion for β-galactosidase (green, single stained neurons=arrows) and SP (red). Double stained neurons are shown by arrowheads, i, SP receptor distribution in the dorsal horn of wild- type mice, j, SP receptor distribution in dorsal horn of Yl7" mice, k, SP receptor staining in lamina I of the contralateral vehicle injected side of Yl7" mice. 1, Loss of cell surface and increase of intracellular SP receptor immunoreactivity in lamina I ten minutes after capsaicin injection into the hindpaw of Yl 7" mice. Scale bar in (e) is 300 μm, in (f), (i) and (j) 80 μm, in (g) and (h) 30 μm, in (k) and (1) 20μm. Figure 2. Cutaneous and visceral nociception of wild-type (black bars) and Yl 7" (white bars) mice in the hot-plate, tail-flick, fonnalin, acetic acid, MgS04, von Frey hair and in neuropathic pain assays as well as in stress and
NPY produced analgesia, a, Latency to shaking of hind-paw or jumping, b Tail-flick latency, c, Mechanical threshold assayed by von Frey hairs, d, Measurement of the number of events (lifting, shaking, licking and biting of the injected paw) in the fonnalin assay. The numbers on the X-axis indicate the concentration in percent of fonnalin administered subcutaneously. e and f, Visceral pain response (abdominal stretching) produced by intraperitoneal injection of diluted acetic acid (e), or MgSθ4 (f). g, stress-induced analgesia in the hot plate assay, h, Development of mechanical allodynia of wild-type and Y l 7" mice in a chronic pain model, i, Analgesic response to tail-flick following an intrathecal injection of NPY. Data are presented as % analgesia. All data are mean ± SEM and statistical analysis was performed by unpaired student's t-test (a-g and i) or two-tailed Mann Whitney U-test (h). *, PO.05; **, P<0.01 ; ***, PO.001.
Figure 3. Neurogenic and non-neurogenic inflammation in wild-type and Yl" '' mice, a, Paws of wild-type and Yl7" mice 30 min after injection of capsaicin (neurogenic inflammation) or vehicle, b, Quantification of evans blue extravasation after capsaicin or vehicle injection, c, Percentage of paw diameter increase of vehicle and capsaicin injected paws, d, Mechanical sensitisation before and after capsaicin-induced inflammation, e and f, Evans blue extravasation (e) and paw diameter (f) 4 hours after carrageenan (non- neurogenic) induced inflammation in the wild-type and Yl 7" mice as indicated, g, Mechanical sensitisation 3 h after carrageenan induced inflammation, h, Quantification of evans blue extravasation of the paws 30 min after mustard oil administration, i, Quantification of evans blue extravasation 30 min after vehicle or SP administration. In all experiments open bars are vehicle control side and black bars the experimental side. All data are mean ± SEM. Statistical analysis was perfonned by unpaired student's t-test. *, P<0.05; **, P<0.01 ; ***, PO.001 .
Figure 4. Measurement of SP release by capsaicin administration in the skin by EIA and effects of Yl agonist and antagonist in inflammation-induced plasma extravasation, a, Released SP in vehicle and capsaicin injected skin, b, Evans blue extravasation 30 min after NPY Yl receptor agonist [Leu31- Pro34]-NPY or vehicle injection intraplantarly. c, Capsaicin-induced evans blue extravasation in wild-type mice in the presence or absence of NPY Yl receptor selective antagonist BIBP 3226. In all experiments open bars are the vehicle control side and black bars the experimental side. All data are mean ± SEM and statistical analysis was performed by unpaired student's t-test. *, P<0.05; **, PO.01 ; ***, PO.001.
References
1 Munglani, R , Hudspith, M J & Hunt, S P The therapeutic potential of neuropeptide Y Analgesic, anxiolytic and antihypertensive Drugs 52, 371 -389 ( 1996)
2 Hua, X Y et al. The antinociceptive effects of spinally administered neuropeptide Y in the rat systematic studies on structure-activity relationship J Pharmacol Exp Ther 258, 243-248 ( 1991 )
3 Duggan, A W , Hope, P J & Lang, C W Microinjection of neuropeptide Y into the superficial dorsal horn reduces stimulus-evoked release of immunoreactive substance P in the anaesthetized cat Neuroscience 44, 733-740 (1 91)
4 Hudspith, M J & Munglani, R Neuropeptide Y- friend or foe Eur J Pain 3, 3-6 (1999)
5 Pedrazzini, T et al. Cardiovascular response, feeding behavior and locomotor activity in mice lacking the NPY Yl receptor Nat Med 4, 722-726 ( 1998)
6 Naveilhan, P , Neveu, I , Arenas, E & Ernfors, P Complementary and overlapping expression of Yl, Y2 and Y5 receptors in the developing and adult mouse nervous system Neuroscience 87, 289-302 ( 1998)
7 Zhang, X et al. Localization of neuropeptide Y Yl receptors in the rat nervous system with special reference to somatic receptors on small dorsal root ganglion neurons Proc NatlAcad Sci USA 91, 1 1738-1 1742 (1994)
8 Snider, W D & McMahon, S B Tackling pain at the source new ideas about nociceptors Neuron 20, 629-632 (1998)
9 Allen, B J et al. Primary afferent fibers that contribute to increased substance P receptor internalization in the spinal cord after injury J Neurophysiol 81, 1379-1390 (1999)
10 Mantyh, P W et al. Receptor endocytosis and dendrite reshaping in spinal neurons after somatosensory stimulation Science 268, 1629-1632 (1995)
1 1 Cao, Y Q et al. Primary afferent tachy inins are required to experience moderate to intense pain Nature 392, 390-394 (1998) 12 Watkins, L R & Mayer, D J Multiple endogenous opiate and non-opiate analgesia systems evidence of their existence and clinical implications Ann N Y Acad Set 467, 273- 299 ( 1986)
13 Rubinstein, M et al. Absence of opioid stress-induced analgesia in mice lacking beta- endorphin by site-directed mutagenesis Proc Natl Acad Set U S A 93, 3995-4000 ( 1996)
14 Marek, P , Mogil, J S , Sternberg, W F , Panocka, I & Liebeskind, J C N-methyl- D-aspartic acid (NMD A) receptor antagonist MK-801 blocks non- opioid stress-induced analgesia II Comparison across three swim- stress paradigms in selectively bred mice Brain Res 578, 197-203 ( 1992)
15 Zhang, X , Wiesenfeld-Hallin, Z & Hokfelt, T Effect of peripheral axotomy on expression of neuropeptide Y receptor mRNA in rat lumbar dorsal root ganglia kur J Neurosct ό, 43-57 ( 1994)
16 Xu, X J , Hao, J X , Hokfelt, T & Wiesenfeld-Hallin, Z The effects of intrathecal neuropeptide Y on the spinal nociceptive flexor reflex in rats with intact sciatic nerves and after peripheral axotomy Neuroscience 63, 817-826 (1994)
17 Jancso, N , Jancso-Gabor, A & Szolcsanyi, J Direct evidence for neurogenic inflammation and its prevention by denervation and by pretreatment with capsaicin Br J Pharmacol 3 1 , 138-151 (1967)
18 Jancso, N , Jancso-Gabor, A & Szolcsanyi, J The role of sensory nerve endings in neurogenic inflammation induced in human skin and in the eye and paw of the rat Br J Pharmacol 33, 32-41 ( 1968)
19 Inoue, H , Asaka, T , Nagata, N & Koshihara, Y Mechanism of mustard oil-induced skin inflammation in mice Eur J Pharamacol 333, 2 1-240 (1997)
20 Semos, M L & Headley, P M The role of nitric oxide in spinal nociceptive reflexes in rats with neurogenic and non-neurogenic peripheral inflammation Neuropharmacology 33, 1487-1497 (1994)
21 Lembeck, F & Holzer, P Substance P as neurogenic mediator of antidromic vasodilation and neurogenic plasma extravasation. Naunyn Schmiedelbergs Arch Pharmacol 3 10, 175-183 (1979) 22 Shi, T J , Zhang, X , Berg, O G , Erickson, J C , Pal iter, R D & Hokflt, T Effect of peripheral axotomy on dorsal root ganglion neuron phenotype and autotomy behaviour in neuropeptide Y-deficient mice Regitl Pept 25, 161 - 173 ( 1998)
23 Wilcox, G L Excitatory neurotransmitters and pain (ed Bond M R , Charlton J R , & Woolf C J ) 97- 1 17 (Elsevier Science Publishers BV, New York, 1991)
24 Walker, M W , Ewald, D A , Perney, T M & Miller, R J Neuropeptide Y modulates neurotransmitter release and Ca2+ currents in rat sensory neurons J Neurosci 8, 2438-2446 ( 1988)
25 Polgar, E , Shehab, S A , Watt, C & Todd, A J GABAergic neurons that contain neuropeptide Y selectively target cells with the neurokinin 1 receptor in laminae III and IV of the rat spinal cord J Neurosci 19, 2637-2646 (1999)
26 Parker, D et al. Co-localized neuropeptide Y and GABA have complementary presynaptic effects on sensory synaptic transmission Eur J Neurosci 10, 2856-2870 ( 1998)
27 De Felipe, C et al. Altered nociception, analgesia and aggression in mice lacking the receptoi for substance P Nature 392, 394-397 (1998)
28 Naveilhan, P et al. Normal feeding behavior, body weight and leptin response require the neuropeptide Y Y2 receptor Nat Me 5, 1 188- 1 193 (1999)
29 Seltzer, Z , Dubner, R & Shir, Y A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury Pain 43, 205-218 ( 1990)
30 Malmberg, A B & Basbaum, A I Partial sciatic nerve injury in the mouse as a model of neuropathic pain behavioral and neuroanatomical correlates Pain 76, 215-222 ( 1998)

Claims (6)

1. Use of a selective neuropeptide Y Y 1 receptor antagonist for preparation of a drug for preventing and/or treating inflammatory conditions.
2. Use according to claim 1, wherein the NPY Yl receptor antagonist is topically, subcutaneously or systemically administered for the treatment of cutaneous inflammation.
3. Use according to claim 1, wherein the NPY Y l receptor antagonist is used topically or systemically for the treatment of internal inflammation.
4. Use according to claim 1, wherein the NPY Y l receptor antagonist is used for treatment of neurogenic inflammation.
5. Use according to one or more of claims 1-4, wherein the NPY Y l receptor is used for treatment of acute and chronic/ persistent inflammation.
6. Use of a NPY Y l receptor as a drug target in screening procedures to find anti-inflammatory compounds.
AU2001248972A 2000-04-13 2001-04-12 Use of npy y1 receptor antagonists in the treatment of inflammatory conditions Abandoned AU2001248972A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
SE0001373 2000-04-13
SE0001373A SE0001373D0 (en) 2000-04-13 2000-04-13 NPY Y1 receptor agonists and antagonists
PCT/SE2001/000828 WO2001078763A1 (en) 2000-04-13 2001-04-12 Use of npy y1 receptor antagonists in the treatment of inflammatory conditions

Publications (1)

Publication Number Publication Date
AU2001248972A1 true AU2001248972A1 (en) 2001-10-30

Family

ID=20279307

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2001248972A Abandoned AU2001248972A1 (en) 2000-04-13 2001-04-12 Use of npy y1 receptor antagonists in the treatment of inflammatory conditions
AU2001248971A Abandoned AU2001248971A1 (en) 2000-04-13 2001-04-12 Use of npy y1 receptor agonists in the treatment of pain conditions

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2001248971A Abandoned AU2001248971A1 (en) 2000-04-13 2001-04-12 Use of npy y1 receptor agonists in the treatment of pain conditions

Country Status (6)

Country Link
US (1) US20030153487A1 (en)
EP (1) EP1276498A1 (en)
AU (2) AU2001248972A1 (en)
CA (1) CA2405600A1 (en)
SE (1) SE0001373D0 (en)
WO (2) WO2001078762A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2262567T3 (en) * 2001-03-20 2006-12-01 Schwarz Pharma Ag NEW USE OF A PEPTIDIC COMPOSITE CLASS FOR THE TREATMENT OF NON-NEUROPATIC INFLAMMATORY PAIN.
DE60100055T2 (en) 2001-03-21 2003-07-24 Sanol Arznei Schwarz Gmbh New use of a class of peptide compounds for the treatment of allodynia or other types of chronic or phantom pain
EP1689378B1 (en) 2003-12-02 2009-04-15 Schwarz Pharma Ag Novel use of peptide compounds for treating central neuropathic pain
EP1734980A1 (en) 2004-04-16 2006-12-27 Schwarz Pharma Ag Use of peptidic compounds for the prophylaxis and treatment of chronic headache
WO2005114213A2 (en) * 2004-05-21 2005-12-01 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with g protein-coupled receptor npy1 (npy1)
EP1604655A1 (en) 2004-06-09 2005-12-14 Schwarz Pharma Ag Novel use of peptide compounds for treating pain in trigeminal neuralgia
CA2573125A1 (en) 2004-08-27 2006-03-02 Schwarz Pharma Ag Novel use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain
EA019757B1 (en) 2006-06-15 2014-06-30 ЮСиБи ФАРМА ГМБХ Pharmaceutical composition with synergistic anticonvulsant effect

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5569742A (en) * 1994-06-22 1996-10-29 The Salk Institute For Biological Studies Centrally truncated NPY cyclic peptides

Also Published As

Publication number Publication date
SE0001373D0 (en) 2000-04-13
AU2001248971A1 (en) 2001-10-30
EP1276498A1 (en) 2003-01-22
US20030153487A1 (en) 2003-08-14
CA2405600A1 (en) 2001-10-25
WO2001078763A1 (en) 2001-10-25
WO2001078762A1 (en) 2001-10-25

Similar Documents

Publication Publication Date Title
Naveilhan et al. Reduced antinociception and plasma extravasation in mice lacking a neuropeptide Y receptor
Eva et al. Physiology and gene regulation of the brain NPY Y1 receptor
Dray et al. New pharmacological strategies for pain relief
Dong et al. Sex-related differences in NMDA-evoked rat masseter muscle afferent discharge result from estrogen-mediated modulation of peripheral NMDA receptor activity
Gupta et al. Potential role of female sex hormones in the pathophysiology of migraine
Arnold et al. Brain renin–angiotensin system in the nexus of hypertension and aging
Li et al. Angiotensin-mediated increase in renal sympathetic nerve discharge within the PVN: role of nitric oxide
Resch et al. Stimulation of the hypothalamic ventromedial nuclei by pituitary adenylate cyclase-activating polypeptide induces hypophagia and thermogenesis
Wang et al. Astroglial modulation of hydromineral balance and cerebral edema
Hay et al. Yes! Sex matters: sex, the brain and blood pressure
Kasper et al. Growth, metabolism, and blood pressure disturbances during aging in transgenic rats with altered brain renin-angiotensin systems
Elmquist Anatomic basis of leptin action in the hypothalamus
Pinilla et al. Characterization of the reproductive effects of the anorexigenic VGF-derived peptide TLQP-21: in vivo and in vitro studies in male rats
Morgan et al. The NPY Y1 receptor antagonist BIBP 3226 blocks NPY induced feeding via a non-specific mechanism
Sun et al. An antinociceptive role of galanin in the arcuate nucleus of hypothalamus in intact rats and rats with inflammation
Kawabe et al. Cardiovascular function of a glutamatergic projection from the hypothalamic paraventricular nucleus to the nucleus tractus solitarius in the rat
US20030153487A1 (en) Use of npy y1 receptor antagonists in the treatment of inflammatory conditions
Samsam Central nervous system acting drugs in treatment of migraine headache
Takizawa et al. Therapeutic implications of cortical spreading depression models in migraine
Ciriello et al. 17β-Estradiol alters the response of subfornical organ neurons that project to supraoptic nucleus to plasma angiotensin II and hypernatremia
Neugebauer et al. Pain-related cortico-limbic plasticity and opioid signaling
Liu et al. EphB receptor signaling in mouse spinal cord contributes to physical dependence on morphine
Iovino et al. Molecular mechanisms involved in the control of neurohypophyseal hormones secretion
Takemura et al. GABAA receptor-mediated effects on expression of c-Fos in rat trigeminal nucleus following high-and low-intensity afferent stimulation
Nagy et al. Control of prolactin secretion by excitatory amino acids