Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Hazard Mater ; 465: 133360, 2024 03 05.
Article in English | MEDLINE | ID: mdl-38157815

ABSTRACT

BACKGROUND: Dibutyl phthalate (DBP), a commonly used plasticizer, has been found to be strongly linked to a consistently high prevalence of allergic diseases, particularly allergic asthma. Previous animal experiments have demonstrated that exposure to DBP can worsen asthma by triggering the production of calcitonin gene-related peptide (CGRP), a neuropeptide in the lung tissue. However, the precise neuroimmune mechanism and pathophysiology of DBP-exacerbated allergic asthma with the assistance of CGRP remain unclear. OBJECTIVE: The present study was to investigate the potential pathophysiological mechanism in DBP-exacerbated asthma from the perspective of neural-immune interactions. METHODS AND RESULTS: C57BL/6 mice were orally exposed to different concentrations (0.4, 4, 40 mg/kg) of DBP for 28 days. They were then sensitized with OVA and nebulized with OVA for 7 consecutive excitations. To investigate whether DBP exacerbates allergic asthma in OVA induced mice, we analyzed airway hyperresponsiveness and lung histopathology. To investigate the activation of JNC and TRPV1 neurons and the release of CGRP by JNC cells, we measured the levels of TRPV1 channels, calcium inward flow, and downstream neuropeptide CGRP. Results showed that TRPV1 expression, inward calcium flux, and CGRP levels were significantly elevated in the lung tissues of the 40DBP + OVA group, suggesting the release of CGRP by JNC cells. To counteract the detrimental effects of DBP mediated by CGRP, we employed olcegepant (also known as BIBN-4096), a CGRP receptor specific antagonist. Results revealed that 40DBP + OVA + olcegepant led to notable decreases in TRPV1, calcium inward flow, and CGRP expression in lung tissues compare with 40DBP + OVA, further supporting the efficacy of olcegepant. Additionally, we also conducted ILC2 flow sorting and observed that neuropeptide CGRP-activated ILC2 cells have a crucial role as key effector cells in DBP-induced neuroimmune positive feedback regulation. Finally, we examined the protein expression of CGRP, GATA3 and P-GATA3, and found that significant upregulations of CGRP and P-GATA3 in the 40DBP + OVA group, suggest that GATA3 acted as a key regulator of CGRP-activated ILC2. CONCLUSION: The aforementioned studies indicate that exposure to DBP can exacerbate allergic asthma, leading to airway inflammation. This exacerbation occurs through the activation of TRPV1 in JNC, resulting in the release of CGRP. The excessive release of CGRP further promotes the release of Th2 cytokines by inducing the activation of ILC2 through GATA phosphorylation. Consequently, this process contributes to the development of airway inflammation and allergic asthma. The increased production of Th2 cytokines also triggers the production of IgE, which interacts with FcεRI on JNC neurons, thereby mediating neuro-immune positive feedback regulation.


Subject(s)
Asthma , Hypersensitivity , Neuropeptides , Mice , Animals , Calcitonin Gene-Related Peptide/toxicity , Calcitonin Gene-Related Peptide/metabolism , Immunity, Innate , Feedback , Dibutyl Phthalate/toxicity , Neuroimmunomodulation , Calcium , Lymphocytes , Mice, Inbred C57BL , Asthma/chemically induced , Asthma/metabolism , Lung/pathology , Cytokines , Neuropeptides/toxicity , Inflammation/pathology , Mice, Inbred BALB C , Ovalbumin
2.
Cephalalgia ; 39(14): 1762-1775, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31550910

ABSTRACT

BACKGROUND: Acute and persistent post-traumatic headache are often debilitating consequences of traumatic brain injury. Underlying physiological mechanisms of post-traumatic headache and its persistence remain unknown, and there are currently no approved therapies for these conditions. Post-traumatic headache often presents with a migraine-like phenotype. As calcitonin-gene related peptide promotes migraine headache, we explored the efficacy and timing of intervention with an anti- calcitonin-gene related peptide monoclonal antibody in novel preclinical models of acute post-traumatic headache and persistent post-traumatic headache following a mild traumatic brain injury event in mice. METHODS: Male, C57Bl/6 J mice received a sham procedure or mild traumatic brain injury resulting from a weight drop that allowed free head rotation while under minimal anesthesia. Periorbital and hindpaw tactile stimulation were used to assess mild traumatic brain injury-induced cutaneous allodynia. Two weeks after the injury, mice were challenged with stress, a common aggravator of migraine and post-traumatic headache, by exposure to bright lights (i.e. bright light stress) and cutaneous allodynia was measured hourly for 5 hours. A murine anti- calcitonin-gene related peptide monoclonal antibody was administered after mild traumatic brain injury at different time points to allow evaluation of the consequences of either early and sustained calcitonin-gene related peptide sequestration or late administration only prior to bright light stress. RESULTS: Mice with mild traumatic brain injury, but not a sham procedure, exhibited both periorbital and hindpaw cutaneous allodynia that resolved by post-injury day 13. Following resolution of injury-induced cutaneous allodynia, exposure to bright light stress re-instated periorbital and hindpaw cutaneous allodynia in injured, but not sham mice. Repeated administration of anti-calcitonin-gene related peptide monoclonal antibody at 2 hours, 7 and 14 days post mild traumatic brain injury significantly attenuated the expression of cutaneous allodynia when evaluated over the 14-day post injury time course and also prevented bright light stress-induced cutaneous allodynia in injured mice. Administration of anti-calcitonin-gene related peptide monoclonal antibody only at 2 hours and 7 days after mild traumatic brain injury blocked injury-induced cutaneous allodynia and partially prevented bright light stress-induced cutaneous allodynia. A single administration of anti-calcitonin-gene related peptide monoclonal antibody after the resolution of the peak injury-induced cutaneous allodynia, but prior to bright light stress challenge, did not prevent bright light stress-induced cutaneous allodynia. CONCLUSIONS: We used a clinically relevant mild traumatic brain injury event in mice along with a provocative stimulus as novel models of acute post-traumatic headache and persistent post-traumatic headache. Following mild traumatic brain injury, mice demonstrated transient periorbital and hindpaw cutaneous allodynia suggestive of post-traumatic headache-related pain and establishment of central sensitization. Following resolution of injury-induced cutaneous allodynia, exposure to bright light stress re-established cutaneous allodynia, suggestive of persistent post-traumatic headache-related pain. Continuous early sequestration of calcitonin-gene related peptide prevented both acute post-traumatic headache and persistent post-traumatic headache. In contrast, delayed anti-calcitonin-gene related peptide monoclonal antibody treatment following establishment of central sensitization was ineffective in preventing persistent post-traumatic headache. These observations suggest that mechanisms involving calcitonin-gene related peptide underlie the expression of acute post-traumatic headache, and drive the development of central sensitization, increasing vulnerability to headache triggers and promoting persistent post-traumatic headache. Early and continuous calcitonin-gene related peptide blockade following mild traumatic brain injury may represent a viable treatment option for post-traumatic headache and for the prevention of post-traumatic headache persistence. ABBREVIATIONS: CA Cutaneous allodynia CGRP Calcitonin gene-related peptide mTBI Mild traumatic brain injury PTH Post-traumatic headache APTH Acute post-traumatic headache PPTH Persistent post-traumatic headache.


Subject(s)
Brain Concussion/chemically induced , Brain Concussion/drug therapy , Calcitonin Gene-Related Peptide Receptor Antagonists/therapeutic use , Calcitonin Gene-Related Peptide/toxicity , Post-Traumatic Headache/chemically induced , Post-Traumatic Headache/drug therapy , Acute Disease , Animals , Brain Concussion/physiopathology , Chronic Disease , Male , Mice , Mice, Inbred C57BL , Post-Traumatic Headache/physiopathology , Vasodilator Agents/toxicity
3.
J Headache Pain ; 19(1): 105, 2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30409109

ABSTRACT

BACKGROUND: Migraine prevention with erenumab and migraine induction by calcitonin gene-related peptide (CGRP) both carry notable individual variance. We wanted to explore a possible association between individual efficacy of anti-CGRP treatment and susceptibility to migraine induction by CGRP. METHODS: Thirteen migraine patients, previously enrolled in erenumab anti-CGRP receptor monoclonal antibody trials, received CGRP in a double-blind, placebo-controlled, randomized cross-over design to investigate their susceptibility to migraine induction. A standardized questionnaire was used to assess the efficacy of previous antibody treatment. The patients were stratified into groups of high responders and poor responders. Primary outcomes were incidence of migraine-like attacks and area under the curve of headache intensity after infusion of CGRP and placebo. All interviews and experiments were performed in laboratories at the Danish Headache Center, Copenhagen, Denmark. RESULTS: Ten high responders and three poor responders were included. CGRP induced migraine-like attacks in ten (77%) patients, whereof two were poor responders, compared to none after placebo (p = 0.002). The area under the curve for headache intensity was greater after CGRP, compared to placebo, at 0-90 min (p = 0.009), and 2-12 h (p = 0.014). The median peak headache intensity score was 5 (5-9) after CGRP, compared to 2 (0-4) after placebo (p = 0.004). CONCLUSIONS: Patients with an excellent effect of erenumab are highly susceptible to CGRP provocation. If an association is evident, CGRP provocation could prove a biomarker for predicting antibody treatment efficacy. TRIAL REGISTRATION: Retrospectively registered at clinicaltrials.gov with identifier: NCT03481400 .


Subject(s)
Antibodies, Monoclonal/therapeutic use , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/toxicity , Migraine Disorders/chemically induced , Migraine Disorders/drug therapy , Adult , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cross-Over Studies , Denmark/epidemiology , Double-Blind Method , Female , Humans , Male , Middle Aged , Migraine Disorders/epidemiology , Retrospective Studies , Young Adult
4.
Pain ; 159(11): 2306-2317, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29994995

ABSTRACT

Migraine is the third most common disease in the world (behind dental caries and tension-type headache) with an estimated global prevalence of 15%, yet its etiology remains poorly understood. Recent clinical trials have heralded the potential of therapeutic antibodies that block the actions of the neuropeptide calcitonin gene-related peptide (CGRP) or its receptor to prevent migraine. Calcitonin gene-related peptide is believed to contribute to trigeminal nerve hypersensitivity and photosensitivity in migraine, but a direct role in pain associated with migraine has not been established. In this study, we report that peripherally administered CGRP can act in a light-independent manner to produce spontaneous pain in mice that is manifested as a facial grimace. As an objective validation of the orbital tightening action unit of the grimace response, we developed a squint assay using a video-based measurement of the eyelid fissure, which confirmed a significant squint response after CGRP injection, both in complete darkness and very bright light. These indicators of discomfort were completely blocked by preadministration of a monoclonal anti-CGRP-blocking antibody. However, the nonsteroidal anti-inflammatory drug meloxicam failed to block the effect of CGRP. Interestingly, an apparent sex-specific response to treatment was observed with the antimigraine drug sumatriptan partially blocking the CGRP response in male, but not female mice. These results demonstrate that CGRP can induce spontaneous pain, even in the absence of light, and that the squint response provides an objective biomarker for CGRP-induced pain that is translatable to humans.


Subject(s)
Calcitonin Gene-Related Peptide/toxicity , Pain/chemically induced , Pain/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies/therapeutic use , Calcitonin Gene-Related Peptide/immunology , Disease Models, Animal , Facial Pain/chemically induced , Facial Pain/drug therapy , Injections, Intraperitoneal , Locomotion/drug effects , Meloxicam , Mice , Mice, Inbred C57BL , Pain/drug therapy , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Sumatriptan/therapeutic use
5.
Purinergic Signal ; 12(3): 561-74, 2016 09.
Article in English | MEDLINE | ID: mdl-27369815

ABSTRACT

Extracellular ATP is suspected to contribute to migraine pain but regulatory mechanisms controlling pro-nociceptive purinergic mechanisms in the meninges remain unknown. We studied the peculiarities of metabolic and signaling pathways of ATP and its downstream metabolites in rat meninges and in cultured trigeminal cells exposed to the migraine mediator calcitonin gene-related peptide (CGRP). Under resting conditions, meningeal ATP and ADP remained at low nanomolar levels, whereas extracellular AMP and adenosine concentrations were one-two orders higher. CGRP increased ATP and ADP levels in meninges and trigeminal cultures and reduced adenosine concentration in trigeminal cells. Degradation rates for exogenous nucleotides remained similar in control and CGRP-treated meninges, indicating that CGRP triggers nucleotide release without affecting nucleotide-inactivating pathways. Lead nitrate-based enzyme histochemistry of whole mount meninges revealed the presence of high ATPase, ADPase, and AMPase activities, primarily localized in the medial meningeal artery. ATP and ADP induced large intracellular Ca(2+) transients both in neurons and in glial cells whereas AMP and adenosine were ineffective. In trigeminal glia, ATP partially operated via P2X7 receptors. ATP, but not other nucleotides, activated nociceptive spikes in meningeal trigeminal nerve fibers providing a rationale for high degradation rate of pro-nociceptive ATP. Pro-nociceptive effect of ATP in meningeal nerves was reproduced by α,ß-meATP operating via P2X3 receptors. Collectively, extracellular ATP, which level is controlled by CGRP, can persistently activate trigeminal nerves in meninges which considered as the origin site of migraine headache. These data are consistent with the purinergic hypothesis of migraine pain and suggest new targets against trigeminal pain.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Migraine Disorders/metabolism , Nucleotides/metabolism , Receptors, Purinergic/metabolism , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcitonin Gene-Related Peptide/toxicity , Cell Separation , Disease Models, Animal , Homeostasis/physiology , Immunohistochemistry , Male , Meninges , Patch-Clamp Techniques , Rats , Rats, Wistar , Trigeminal Ganglion/metabolism
6.
J Neurosci ; 34(1): 60-5, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24381268

ABSTRACT

The bed nucleus of the stria terminalis (BNST) is thought to generate anxiety-like states via its projections to autonomic and neuroendocrine regulatory structures of the brain. However, because most BNST cells are GABAergic, they are expected to inhibit target neurons. In contrast with this, infusion of calcitonin gene-related peptide (CGRP) into BNST was reported to potentiate anxiety while activating BNST targets. The present study aimed to shed light on this paradox. The CGRP innervation of BNST originates in the pontine parabrachial nucleus and targets its anterolateral sector (BNST-AL). Thus, we investigated the effects of CGRP on BNST-AL neurons using patch recordings in vitro in male rats. CGRP did not alter the passive properties of BNST-AL cells but increased the amplitude of IPSPs evoked by stimulation of the stria terminalis (ST). However, IPSP paired-pulse ratios were unchanged by CGRP, and there was no correlation between IPSP potentiation and variance, suggesting that CGRP acts postsynaptically. Consistent with this, CGRP hyperpolarized the GABA-A reversal of BNST-AL cells. These results indicate that CGRP increases ST-evoked GABA-A IPSPs and hyperpolarizes their reversal potential through a postsynaptic change in Cl(-) homeostasis. Overall, our findings suggest that CGRP potentiates anxiety-like behaviors and increases neural activity in BNST targets, by inhibiting BNST-AL cells, supporting the conclusion that BNST-AL exerts anxiolytic effects.


Subject(s)
Anxiety/physiopathology , Calcitonin Gene-Related Peptide/toxicity , Fear/physiology , Neural Inhibition/physiology , Neurons/physiology , Septal Nuclei/physiology , Animals , Anxiety/chemically induced , Anxiety/psychology , Fear/drug effects , Fear/psychology , Male , Neural Inhibition/drug effects , Neurons/drug effects , Organ Culture Techniques , Rats , Rats, Inbred Lew , Septal Nuclei/drug effects
7.
Exp Brain Res ; 196(1): 179-93, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19288089

ABSTRACT

Migraine is a complex neurological disease with a genetic background. Headache is the most prominent and clinically important symptom of migraine but its origin is still enigmatic. Numerous clinical, histochemical, electrophysiological, molecular and genetical approaches form a puzzle of findings that slowly takes shape. The generation of primary headaches like migraine pain seems to be the consequence of multiple pathophysiological changes in meningeal tissues, the trigeminal ganglion, trigeminal brainstem nuclei and descending inhibitory systems, based on specific characteristics of the trigeminovascular system. This contribution reviews the current discussion of where and how the migraine pain may originate and outlines the experimental work to answer these questions.


Subject(s)
Headache/etiology , Headache/physiopathology , Migraine Disorders/physiopathology , Pain/etiology , Pain/physiopathology , Animals , Calcitonin Gene-Related Peptide/toxicity , Histamine/toxicity , Humans , Meninges/physiopathology , Migraine Disorders/complications , Models, Neurological , Neural Inhibition/physiology , Neurons/physiology , Nitrogen Compounds/toxicity , Trigeminal Ganglion/physiopathology , Trigeminal Nuclei/physiopathology
8.
Mol Pain ; 3: 30, 2007 Oct 25.
Article in English | MEDLINE | ID: mdl-17961222

ABSTRACT

BACKGROUND: Acute cutaneous neurogenic inflammation initiated by activation of transient receptor potential vanilloid-1 (TRPV1) receptors following intradermal injection of capsaicin is mediated mainly by dorsal root reflexes (DRRs). Inflammatory neuropeptides are suggested to be released from primary afferent nociceptors participating in inflammation. However, no direct evidence demonstrates that the release of inflammatory substances is due to the triggering of DRRs and how activation of TRPV1 receptors initiates neurogenic inflammation via triggering DRRs. RESULTS: Here we used pharmacological manipulations to analyze the roles of TRPV1 and neuropeptidergic receptors in the DRR-mediated neurogenic inflammation induced by intradermal injection of capsaicin. The degree of cutaneous inflammation in the hindpaw that followed capsaicin injection was assessed by measurements of local blood flow (vasodilation) and paw-thickness (edema) of the foot skin in anesthetized rats. Local injection of capsaicin, calcitonin gene-related peptide (CGRP) or substance P (SP) resulted in cutaneous vasodilation and edema. Removal of DRRs by either spinal dorsal rhizotomy or intrathecal administration of the GABAA receptor antagonist, bicuculline, reduced dramatically the capsaicin-induced vasodilation and edema. In contrast, CGRP- or SP-induced inflammation was not significantly affected after DRR removal. Dose-response analysis of the antagonistic effect of the TRPV1 receptor antagonist, capsazepine administered peripherally, shows that the capsaicin-evoked inflammation was inhibited in a dose-dependent manner, and nearly completely abolished by capsazepine at doses between 30-150 mug. In contrast, pretreatment of the periphery with different doses of CGRP8-37 (a CGRP receptor antagonist) or spantide I (a neurokinin 1 receptor antagonist) only reduced the inflammation. If both CGRP and NK1 receptors were blocked by co-administration of CGRP8-37 and spantide I, a stronger reduction in the capsaicin-initiated inflammation was produced. CONCLUSION: Our data suggest that 1) the generation of DRRs is critical for driving the release of neuropeptides antidromically from primary afferent nociceptors; 2) activation of TRPV1 receptors in primary afferent nociceptors following intradermal capsaicin injection initiates this process; 3) the released CGRP and SP participate in neurogenic inflammation.


Subject(s)
Neurogenic Inflammation/etiology , Receptors, Neurokinin-1/physiology , Reflex , Spinal Nerve Roots/physiopathology , TRPV Cation Channels/physiology , Analgesics/pharmacology , Animals , Bicuculline/pharmacology , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide/toxicity , Capsaicin/administration & dosage , Capsaicin/analogs & derivatives , Capsaicin/toxicity , Edema/chemically induced , Edema/etiology , GABA Antagonists/pharmacology , Injections, Intradermal , Neurogenic Inflammation/chemically induced , Peptide Fragments/pharmacology , Rats , Receptors, Neurokinin-1/agonists , Spinal Nerve Roots/drug effects , Substance P/analogs & derivatives , Substance P/metabolism , Substance P/pharmacology , Substance P/toxicity , TRPV Cation Channels/agonists , Vasodilation/drug effects
9.
Br J Pharmacol ; 125(6): 1304-12, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9863661

ABSTRACT

The contribution of calcitonin gene-related peptide (CGRP) to bilateral oedema formation in the rat hindpaw following an unilateral challenge with CGRP was investigated. Rats were injected into the left hindpaw with either saline, CGRP or a CGRP antagonist (CGRP8-37). All injections were given in a double blind fashion and in a volume of 100 microl. CGRP and CGRP8-37 were administered in concentrations of 75, 150 or 300 pmol. Volumes of the right and left hindpaw were measured every hour for 5 h by plethysmometry. Injection of CGRP 300 pmol into the left hindpaw resulted in a bilaterally increased hindpaw volume after 5 h as compared with the groups given saline. No changes were found in hindpaw volumes following the injection of either 75 or 150 pmol of CGRP or 75, 150 or 300 pmol of CGRP8-37 as compared with saline injection. To elucidate whether or not the bilateral oedema formation was related to a release of endogenous CGRP, microdialysis of the contralateral hindpaw was carried out, and concentrations of CGRP-like immunoreactivity (-LI) were determined by radioimmunoassay and high performance liquid chromatography. Injection of CGRP 300 pmol into the left hindpaw increased the release of CGRP-LI into the right hindpaw perfusate after 4 and 5 h. No changes in CGRP-LI were detected in the right hindpaw perfusate following challenge with saline or CGRP8-37. To study the contribution of the nervous system to the contralateral release of CGRP-LI, sciatic nerve ligated and intact sham-operated rats were used. Sciatic nerve ligation but not sham-operation on the non-injected side abolished the increased release of CGRP-LI following contralateral administration of CGRP 300 pmol. To study the spinal cord mechanisms resulting in the bilateral oedema formation following unilateral challenge with 300 pmol of CGRP, intrathecal pretreatment with either 10 nmol bicuculline (GABA(A) receptor antagonist) or 10 nmol CGRP8-37 was carried out. Bicuculline but not CGRP8-37 abolished the bilateral oedema formation induced by CGRP 300 pmol. In order to study the mechanisms by which administration of CGRP 300 pmol induces oedema, CGRP 300 pmol was administered concomitantly with either 300 pmol of CGRP8-37 (CGRP receptor antagonist), or 3 nmol of promethazine (H1 receptor antagonist), or 3 nmol of s(-)-propranolol (5-HT1 receptor antagonist), or 3 nmol of cyproheptadine (5-HT2 receptor antagonist) or 3 nmol of ICS 205-930 (5-HT3 receptor antagonist). Oedema formation was measured at 1, 5, 7 and 24 h. Injection of CGRP 300 pmol into the left hindpaw induced a bilateral oedema formation which was still significant at 24 h. Concomitant administration of either CGRP8-37, ICS 205-920 or cyproheptadine blocked the oedema formation at 24 h. No effect on oedema formation was found when CGRP 300 pmol was co-administered with either promethazine or s(-)-propranolol (H1 and 5-HT1 receptor antagonists, respectively). The results of the present study show that both the nervous system and local inflammatory processes contribute to bilateral hindpaw oedema formation following unilateral challenge with CGRP 300 pmol. Our results indicate that endogenous release of CGRP following inflammatory response may play an important role in inducing oedema formation.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/toxicity , Edema/chemically induced , Animals , Bicuculline/pharmacology , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Capsaicin/pharmacology , Chromatography, High Pressure Liquid , Drug Interactions , GABA Antagonists/pharmacology , Hindlimb/drug effects , Histamine Antagonists/pharmacology , Humans , Male , Miotics/pharmacology , Peptide Fragments/pharmacology , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology , Thiorphan/pharmacology
10.
Regul Pept ; 71(1): 1-7, 1997 Jul 23.
Article in English | MEDLINE | ID: mdl-9299635

ABSTRACT

We have reported previously that calcitonin gene-related peptide (CGRP) exerts hypertrophic effects, defined in the broadest sense as increased mass of protein per cell, in adult rat ventricular in vitro. The aim of the present investigation was to determine whether the peptide also increases the cell surface area of, and induces expression of ANP and skeletal alpha-actin mRNA in hypertrophying neonatal rat ventricular cardiomyocytes. Cells cultured in the presence of CGRP were invisibly hypertrophied after 48 h compared to cells cultured in serum-free MEM for the same period. CGRP, 100 pM and 1 nM, increased cell surface area significantly and to values 1.82- and 2.15-fold greater, respectively, than in the absence of peptide (659.64 +/-23.48 microns 2, n = 10). The selective antagonist at CGRP1, receptors, CGRP8-37(200nM), significantly attenuated the effects of CGRP (100 pM and 1 nM). CGRP caused a marked up-regulation of the expression of mRNA encoding skeletal alpha-actin and ANP, respectively, maximally after 12 h and at a concentration of 100 pM, to values approximately 3.6- and 2.5-fold greater than in the absence of peptide. These effects of the peptide were completely abolished in the presence of CGRP8-37(100 nM). In conclusion, CGRP increases cell surface area and induces expression of ANP and skeletal alpha-actin mRNA in hypertrophying cardiomyocytes via the CGRP1, receptor subtype.


Subject(s)
Actins/biosynthesis , Atrial Natriuretic Factor/biosynthesis , Calcitonin Gene-Related Peptide/toxicity , Cardiomegaly/chemically induced , Myocardium/cytology , Actins/genetics , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Blotting, Northern , Calcitonin Gene-Related Peptide/pharmacology , Cardiomegaly/physiopathology , Cell Size/drug effects , Cells, Cultured , Culture Media, Serum-Free , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Humans , Miotics/pharmacology , Peptide Fragments/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Calcitonin Gene-Related Peptide/drug effects , Receptors, Calcitonin Gene-Related Peptide/metabolism , Up-Regulation
11.
Arch Int Pharmacodyn Ther ; 330(3): 332-44, 1995.
Article in English | MEDLINE | ID: mdl-8836452

ABSTRACT

The pharmacological characteristics of DQ-2511, a substituted benzamide (3-[[[2-(3,4-dimethoxyphenyl)ethyl]carbamoyl]methyl] amino-N-methylbenzamide), as a prokinetic agent were studied. Cholecystokinin-octapeptide, dopamine, and alpha-calcitonin gene-related peptide, all suppressed gastric emptying in mice. Reversal of the depressed emptying occurred when DQ-2511 was administered by the oral or intraperitoneal route. When the action of eight proposed metabolites of DQ-2511 on the mouse cholecystokinin-octapeptide model was investigated, the main metabolite in plasma, MA-2, showed no effect, although two minor metabolites ameliorated or aggravated the delayed gastric emptying. This finding implies that DQ-2511, as the parent compound itself, exerts the ameliorative action. In dogs treated with cisplatin or copper sulfate, DQ-2511 had no antiemetic activity, as assessed by the number of vomiting episodes. The concern that the mechanism of action of DQ-2511 was blockade of receptors for cholecystokinin-octapeptide, dopamine, serotonin, alpha-calcitonin gene-related peptide, nicotine or muscarine, was resolved by results of radioligand binding studies showing the absence of a DQ-2511 binding to any of these receptor types. Evidence is accumulating that the mechanism of the prokinetic action of DQ-2511 involves the intrinsic and extrinsic autonomic innervation.


Subject(s)
Anti-Ulcer Agents/pharmacology , Benzamides/pharmacology , Gastric Emptying/drug effects , Piperidines/pharmacology , Administration, Oral , Analysis of Variance , Animals , Anti-Ulcer Agents/blood , Benzamides/administration & dosage , Benzamides/blood , Benzamides/therapeutic use , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/toxicity , Cisapride , Cisplatin/toxicity , Copper Sulfate/toxicity , Dogs , Dopamine/metabolism , Dopamine/toxicity , Female , Injections, Intraperitoneal , Male , Mice , Muscarine/metabolism , Muscarine/toxicity , Nicotine/metabolism , Nicotine/toxicity , Piperidines/administration & dosage , Piperidines/therapeutic use , Radioligand Assay , Sincalide/metabolism , Sincalide/toxicity , Structure-Activity Relationship , Vomiting/drug therapy
12.
Inflamm Res ; 44(3): 125-30, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7552577

ABSTRACT

We have investigated the effects of actinomycin D on mouse ear oedema induced by capsaicin, neuropeptides, and established inflammatory mediators. Actinomycin D (0.5 mg/kg, i.v.) significantly (P < 0.01) inhibited ear oedema induced by topical application of capsaicin, while adriamycin (6.0 mg/kg, i.v.) and cycloheximide (6.0 mg/kg, i.v.) had no effect on oedema. The ear oedema induced by intradermal injection of neuropeptides such as mammalian tachykinins, calcitonin gene-related peptide (CGRP), and vasoactive intestinal peptide (VIP), was markedly (P < 0.05, P < 0.01 or P < 0.001) suppressed by actinomycin D. The drug was also effective (P < 0.01 or P < 0.001) in inhibiting bradykinin (BK)- and compound 48/80-induced ear oedema, but did not inhibit oedema induced by histamine, 5-HT, leukotriene C4 (LTC4), and platelet activating factor (PAF) at a dose of 1 mg/kg. In mast cell-deficient W/WV mice, actinomycin D (1.0 mg/kg, i.v.) failed to inhibit substance P (SP)-induced ear oedema whereas spantide (0.5 mg/kg, i.v.) was an effective (P < 0.01) inhibitor of oedema formation. Furthermore, actinomycin D (10-100 microM) dose-dependently prevented histamine release from rat peritoneal mast cells evoked by SP, compound 48/80, and the ionophore A23182, respectively. These results strongly suggest that an inhibitory effect of actinomycin D on neurogenic inflammation is due primarily to the prevention of mast cell activation mediated by neuropeptides, rather than an interaction with DNA or receptors of neuropeptides.


Subject(s)
Dactinomycin/therapeutic use , Edema/drug therapy , Animals , Bradykinin/administration & dosage , Bradykinin/toxicity , Calcimycin/administration & dosage , Calcimycin/toxicity , Calcitonin Gene-Related Peptide/administration & dosage , Calcitonin Gene-Related Peptide/toxicity , Capsaicin/administration & dosage , Capsaicin/toxicity , Cycloheximide/administration & dosage , Cycloheximide/toxicity , Dactinomycin/administration & dosage , Dactinomycin/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Ear Diseases/drug therapy , Edema/chemically induced , Histamine/administration & dosage , Histamine/metabolism , Histamine/toxicity , Injections, Intravenous , Leukotriene C4/administration & dosage , Leukotriene C4/toxicity , Male , Mast Cells/cytology , Mast Cells/drug effects , Mice , Platelet Activating Factor/administration & dosage , Platelet Activating Factor/toxicity , Rats , Rats, Wistar , Serotonin/administration & dosage , Serotonin/toxicity , Substance P/administration & dosage , Substance P/toxicity , Tachykinins/administration & dosage , Tachykinins/toxicity , Vasoactive Intestinal Peptide/administration & dosage , Vasoactive Intestinal Peptide/toxicity , p-Methoxy-N-methylphenethylamine/administration & dosage , p-Methoxy-N-methylphenethylamine/toxicity
13.
Ann N Y Acad Sci ; 657: 412-9, 1992 Jun 30.
Article in English | MEDLINE | ID: mdl-1637097

ABSTRACT

Calcitonin gene-related peptide produces dose-related vasodilatation after intradermal injection in several species. In the present study, CGRP increased blood flow in rabbit skin but had no direct effect on edema formation in rat or rabbit skin or in the rat knee joint. However, CGRP produced significant potentiation of edema formation when co-injected with histamine, a potent mediator of increased vascular permeability. Therefore, release of CGRP from stimulated C-fiber nerves may contribute to the vascular changes that are an integral part of the inflammatory process. The activity of the putative CGRP antagonist CGRP8-37 (300 pmol) against CGRP was also investigated in rabbit and rat skin. Whereas it was found to selectively antagonize the effects of CGRP in rabbit skin, the antagonist produced edema in rat skin at the same dose. Thus, CGRP8-37 may be used in the rabbit to study the effects of endogenously released CGRP, but caution is required when this antagonist is used in the rat.


Subject(s)
Calcitonin Gene-Related Peptide/toxicity , Joint Diseases/chemically induced , Skin Diseases/chemically induced , Skin/blood supply , Animals , Edema , Histamine/pharmacology , Humans , Inflammation , Iodine Radioisotopes , Joint Diseases/physiopathology , Male , Rats , Rats, Inbred Strains , Regional Blood Flow/drug effects , Serum Albumin/metabolism , Skin Diseases/physiopathology , Vasodilation/drug effects , Xenon Radioisotopes
14.
J Biol Chem ; 266(17): 11234-6, 1991 Jun 15.
Article in English | MEDLINE | ID: mdl-2040631

ABSTRACT

Blood feeding by the sand fly Lutzomyia longipalpis is aided by the presence of a vasodilator in its salivary glands. This novel vasodilator has been isolated by reversed-phase high-performance liquid chromatography. Ten nanograms of the vasodilator are present in the extract of a pair of sand fly salivary glands. It has 500 times the vasodilatory activity of calcitonin gene-related peptide, previously the most potent vasodilator peptide known. This novel peptide is thus called maxadilan.


Subject(s)
Insect Hormones/isolation & purification , Insect Proteins , Salivary Glands/chemistry , Amino Acids/analysis , Animals , Calcitonin Gene-Related Peptide/toxicity , Chromatography, High Pressure Liquid , Diptera , Electrophoresis , Electrophoresis, Polyacrylamide Gel , Erythema/chemically induced , Insect Hormones/chemistry , Insect Hormones/toxicity , Isoelectric Focusing , Mass Spectrometry , Rabbits , Recombinant Proteins/toxicity , Vasodilator Agents/isolation & purification
15.
Neurosci Lett ; 124(1): 49-51, 1991 Mar 11.
Article in English | MEDLINE | ID: mdl-1713317

ABSTRACT

The effect of neurokinin A (NKA), substance P (SP) and calcitonin gene-related peptide (CGRP) in peripheral hyperalgesia was studied in rats using a modification of the Randall-Selitto paw test. NKA was 10 times more potent than SP which was 500 times more potent than CGRP in inducing hyperalgesia in the rat paw, suggesting that NKA and SP but not CGRP could have an important role in acute hyperalgesic conditions. Furthermore, sensitization induced by several injections of subthreshold doses of NKA or CGRP suggest that these neuropeptides along with SP could participate as mediators or modulators of chronic pain.


Subject(s)
Calcitonin Gene-Related Peptide/toxicity , Hyperalgesia/chemically induced , Neurokinin A/toxicity , Substance P/toxicity , Animals , Inflammation , Male , Nociceptors/drug effects , Nociceptors/physiology , Rats , Rats, Inbred Strains , Substance P/analogs & derivatives , Substance P/antagonists & inhibitors , Substance P/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...