1
|
Salib AMN, Crane MJ, Jamieson AM, Lipscombe D. Peripheral Ca V2.2 Channels in the Skin Regulate Prolonged Heat Hypersensitivity during Neuroinflammation. eNeuro 2024; 11:ENEURO.0311-24.2024. [PMID: 39433408 PMCID: PMC11599794 DOI: 10.1523/eneuro.0311-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Neuroinflammation can lead to chronic maladaptive pain affecting millions of people worldwide. Neurotransmitters, cytokines, and ion channels are implicated in neuroimmune cell signaling, but their roles in specific behavioral responses are not fully elucidated. Voltage-gated CaV2.2 channel activity in skin controls rapid and transient heat hypersensitivity induced by intradermal (i.d.) capsaicin via IL-1ɑ cytokine signaling. CaV2.2 channels are not, however, involved in mechanical hypersensitivity that developed in the i.d. capsaicin animal model. Here, we show that CaV2.2 channels are also critical for heat hypersensitivity induced by i.d. complete Freund adjuvant (CFA). i.d. CFA, a model of chronic neuroinflammation, involves ongoing cytokine signaling for days leading to pronounced edema and hypersensitivity to sensory stimuli. Peripheral CaV2.2 channel activity in the skin was required for the full development and week-long time course of heat hypersensitivity induced by i.d. CFA, but paw edema and mechanical hypersensitivity were independent of CaV2.2 channel activity. CFA induced increases in several cytokines in hindpaw fluid including IL-6 which was also dependent on CaV2.2 channel activity. Using IL-6-specific neutralizing antibodies in vivo, we show that IL-6 contributes to heat hypersensitivity and that neutralizing both IL-1ɑ and IL-6 was even more effective at reducing the magnitude and duration of CFA-induced heat hypersensitivity. Our findings demonstrate a functional link between CaV2.2 channel activity and the release of IL-6 in the skin and show that CaV2.2 channels have a privileged role in the induction and maintenance of heat hypersensitivity during chronic forms of neuroinflammation in the skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Departments of Neuroscience, Brown University, Providence, Rhode Island 02912
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Meredith J Crane
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Amanda M Jamieson
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Diane Lipscombe
- Departments of Neuroscience, Brown University, Providence, Rhode Island 02912
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
2
|
Salib AMN, Crane MJ, Jamieson AM, Lipscombe D. Peripheral Ca V2.2 channels in skin regulate prolonged heat hypersensitivity during neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603149. [PMID: 39071304 PMCID: PMC11275762 DOI: 10.1101/2024.07.13.603149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Neuroinflammation can lead to chronic maladaptive pain affecting millions of people worldwide. Neurotransmitters, cytokines, and ion channels are implicated in neuro-immune cell signaling but their roles in specific behavioral responses are not fully elucidated. Voltage-gated CaV2.2 channel activity in skin controls rapid and transient heat hypersensitivity induced by intradermal capsaicin via IL-1α cytokine signaling. CaV2.2 channels are not, however, involved in mechanical hypersensitivity that developed in the same animal model. Here, we show that CaV2.2 channels are also critical for heat hypersensitivity induced by the intradermal (id) Complete Freund's Adjuvant (CFA) model of chronic neuroinflammation that involves ongoing cytokine signaling for days. Ongoing CFA-induced cytokine signaling cascades in skin lead to pronounced edema, and hypersensitivity to sensory stimuli. Peripheral CaV2.2 channel activity in skin is required for the full development and week-long time course of heat hypersensitivity induced by id CFA. CaV2.2 channels, by contrast, are not involved in paw edema and mechanical hypersensitivity. CFA induced increases in cytokines in hind paws including IL-6 which was dependent on CaV2.2 channel activity. Using IL-6 specific neutralizing antibodies, we show that IL-6 contributes to heat hypersensitivity and, neutralizing both IL-1α and IL-6 was even more effective at reducing the magnitude and duration of CFA-induced heat hypersensitivity. Our findings demonstrate a functional link between CaV2.2 channel activity and the release of IL-6 in skin and show that CaV2.2 channels have a privileged role in the induction and maintenance of heat hypersensitivity during chronic forms of neuroinflammation in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience & the Carney Institute for Brain Science Brown University, Providence, RI 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience & the Carney Institute for Brain Science Brown University, Providence, RI 02912, USA
| |
Collapse
|
3
|
Salib AMN, Crane MJ, Lee SH, Wainger BJ, Jamieson AM, Lipscombe D. Interleukin-1α links peripheral Ca V2.2 channel activation to rapid adaptive increases in heat sensitivity in skin. Sci Rep 2024; 14:9051. [PMID: 38643253 PMCID: PMC11032389 DOI: 10.1038/s41598-024-59424-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
Neurons have the unique capacity to adapt output in response to changes in their environment. Within seconds, sensory nerve endings can become hypersensitive to stimuli in response to potentially damaging events. The underlying behavioral response is well studied, but several of the key signaling molecules that mediate sensory hypersensitivity remain unknown. We previously discovered that peripheral voltage-gated CaV2.2 channels in nerve endings in skin are essential for the rapid, transient increase in sensitivity to heat, but not to mechanical stimuli, that accompanies intradermal capsaicin. Here we report that the cytokine interleukin-1α (IL-1α), an alarmin, is necessary and sufficient to trigger rapid heat and mechanical hypersensitivity in skin. Of 20 cytokines screened, only IL-1α was consistently detected in hind paw interstitial fluid in response to intradermal capsaicin and, similar to behavioral sensitivity to heat, IL-1α levels were also dependent on peripheral CaV2.2 channel activity. Neutralizing IL-1α in skin significantly reduced capsaicin-induced changes in hind paw sensitivity to radiant heat and mechanical stimulation. Intradermal IL-1α enhances behavioral responses to stimuli and, in culture, IL-1α enhances the responsiveness of Trpv1-expressing sensory neurons. Together, our data suggest that IL-1α is the key cytokine that underlies rapid and reversible neuroinflammatory responses in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Sang Hun Lee
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Brian J Wainger
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
4
|
Salib AMN, Crane MJ, Lee SH, Wainger BJ, Jamieson AM, Lipscombe D. Interleukin-1α links peripheral Ca V2.2 channel activation to rapid adaptive increases in heat sensitivity in skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.17.572072. [PMID: 38585803 PMCID: PMC10996502 DOI: 10.1101/2023.12.17.572072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Neurons have the unique capacity to adapt output in response to changes in their environment. Within seconds, sensory nerve endings can become hypersensitive to stimuli in response to potentially damaging events. The underlying behavioral response is well studied, but several of the key signaling molecules that mediate sensory hypersensitivity remain unknown. We previously discovered that peripheral voltage-gated CaV2.2 channels in nerve endings in skin are essential for the rapid, transient increase in sensitivity to heat, but not to mechanical stimuli, that accompanies intradermal capsaicin. Here we report that the cytokine interleukin-1α (IL-1α), an alarmin, is necessary and sufficient to trigger rapid heat and mechanical hypersensitivity in skin. Of 20 cytokines screened, only IL-1α was consistently detected in hind paw interstitial fluid in response to intradermal capsaicin and, similar to behavioral sensitivity to heat, IL-1α levels were also dependent on peripheral CaV2.2 channel activity. Neutralizing IL-1α in skin significantly reduced capsaicin-induced changes in hind paw sensitivity to radiant heat and mechanical stimulation. Intradermal IL-1α enhances behavioral responses to stimuli and, in culture, IL-1α enhances the responsiveness of Trpv1-expressing sensory neurons. Together, our data suggest that IL-1α is the key cytokine that underlies rapid and reversible neuroinflammatory responses in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Sang Hun Lee
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Brian J Wainger
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| |
Collapse
|
5
|
Ramzan F, Kiran L, Malik SN, Malik MI. Tachykinins Play a Major Role in Micro and Macrovascular Complications in Type 2 Diabetes Patients. Curr Diabetes Rev 2024; 20:e050523216590. [PMID: 37151064 DOI: 10.2174/1573399819666230505123844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 05/09/2023]
Abstract
Diabetes Mellitus is a metabolic disorder, which is characterized by an increase in blood glucose levels. The defects in the secretion or action of insulin are the major cause of diabetes. Increase in the blood glucose level exerts a negative effect on the normal functions of the body organs and this leads to the dysfunctions of cells and tissue and causes vascular complications in diabetic patients. Several studies indicate that neuropeptides are released from the neurosensory cells which are mainly known as tachykinins which provoke major vascular complications in diabetic patients. Tachykinins are known as pro-inflammatory peptides which increase vascular complications and vascular permeability. The duration and severity of diabetes disease increase the risk of vascular complication in patients. The aim of this review is to elaborate the role of tachykinins in microvascular and macrovascular complications in diabetic patients. The study concluded that tachykinins increase micro and macrovascular complications in diabetic patients.
Collapse
Affiliation(s)
- Faiqah Ramzan
- Gomal Centre of Biochemistry and Biotechnology, Gomal University, 29050, Dera Ismail Khan, Pakistan
- Faculty of Veterinary and Animal Sciences, Gomal University, Dera Ismail Khan, Pakistan
| | - Laila Kiran
- Gomal Centre of Biochemistry and Biotechnology, Gomal University, 29050, Dera Ismail Khan, Pakistan
| | - Shumaila Noreen Malik
- Department of Gynecology and Obstetrics, District Teaching Hospital, Dera Ismail Khan, Pakistan
| | | |
Collapse
|
6
|
Safwat A, Helmy A, Gupta A. The Role of Substance P Within Traumatic Brain Injury and Implications for Therapy. J Neurotrauma 2023; 40:1567-1583. [PMID: 37132595 DOI: 10.1089/neu.2022.0510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
This review examines the role of the neuropeptide substance P within the neuroinflammation that follows traumatic brain injury. It examines it in reference to its preferential receptor, the neurokinin-1 receptor, and explores the evidence for antagonism of this receptor in traumatic brain injury with therapeutic intent. Expression of substance P increases following traumatic brain injury. Subsequent binding to the neurokinin-1 receptor results in neurogenic inflammation, a cause of deleterious secondary effects that include an increased intracranial pressure and poor clinical outcome. In several animal models of TBI, neurokinin-1 receptor antagonism has been shown to reduce brain edema and the resultant rise in intracranial pressure. A brief overview of the history of substance P is presented, alongside an exploration into the chemistry of the neuropeptide with a relevance to its functions within the central nervous system. This review summarizes the scientific and clinical rationale for substance P antagonism as a promising therapy for human TBI.
Collapse
Affiliation(s)
- Adam Safwat
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Adel Helmy
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Arun Gupta
- Neurosciences Critical Care Unit, Addenbrooke's Hospital, Cambridge, United Kingdom
| |
Collapse
|
7
|
Perineural Capsaicin Treatment Inhibits Collateral Sprouting of Intact Cutaneous Nociceptive Afferents. Biomedicines 2022; 10:biomedicines10061326. [PMID: 35740347 PMCID: PMC9220090 DOI: 10.3390/biomedicines10061326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Perineural treatment of peripheral nerves with capsaicin produces a long-lasting selective regional thermo- and chemo-analgesia and elimination of the neurogenic inflammatory response involving degeneration of nociceptive afferent fibers. In this study, we examined longitudinal changes in mustard oil–induced sensory neurogenic vasodilatation and plasma extravasation following perineural capsaicin treatment of the rat saphenous nerve utilizing scanning laser Doppler imaging and vascular labeling with colloidal silver. Capsaicin treatment resulted in a marked decrease in mustard oil–induced vasodilatation in the skin area served by the saphenous nerve. Repeated imaging of the vasodilatatory response showed no recovery for at least 7 weeks. However, following transection and ligation of the capsaicin-treated saphenous nerve, a substantial recovery of the vasodilatatory response was observed, suggesting a reinnervation of the chemodenervated skin area by collateral sprouting of neighboring intact sciatic nerve afferents. Elimination of the recovered vascular reaction by capsaicin treatment of the sciatic nerve supported this conclusion. Similar results have been obtained by using the vascular labeling technique. These findings indicate an inhibitory effect of persisting cutaneous nerve fibers on the collateral sprouting of intact nerve fibers into the chemodenervated skin area. These observations may bear implications for the development of sensory disturbances following peripheral nerve injuries.
Collapse
|
8
|
Heat Shock Proteins Alterations in Rheumatoid Arthritis. Int J Mol Sci 2022; 23:ijms23052806. [PMID: 35269948 PMCID: PMC8911505 DOI: 10.3390/ijms23052806] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory and autoimmune disease characterized by the attack of the immune system on the body's healthy joint lining and degeneration of articular structures. This disease involves an increased release of inflammatory mediators in the affected joint that sensitize sensory neurons and create a positive feedback loop to further enhance their release. Among these mediators, the cytokines and neuropeptides are responsible for the crippling pain and the persistent neurogenic inflammation associated with RA. More importantly, specific proteins released either centrally or peripherally have been shown to play opposing roles in the pathogenesis of this disease: an inflammatory role that mediates and increases the severity of inflammatory response and/or an anti-inflammatory and protective role that modulates the process of inflammation. In this review, we will shed light on the neuroimmune function of different members of the heat shock protein (HSPs) family and the complex manifold actions that they exert during the course of RA. Specifically, we will focus our discussion on the duality in the mechanism of action of Hsp27, Hsp60, Hsp70, and Hsp90.
Collapse
|
9
|
Ashina M, Hansen JM, Do TP, Melo-Carrillo A, Burstein R, Moskowitz MA. Migraine and the trigeminovascular system-40 years and counting. Lancet Neurol 2019; 18:795-804. [PMID: 31160203 DOI: 10.1016/s1474-4422(19)30185-1] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/28/2019] [Accepted: 04/04/2019] [Indexed: 12/15/2022]
Abstract
The underlying causes of migraine headache remained enigmatic for most of the 20th century. In 1979, The Lancet published a novel hypothesis proposing an integral role for the neuropeptide-containing trigeminal nerve. This hypothesis led to a transformation in the migraine field and understanding of key concepts surrounding migraine, including the role of neuropeptides and their release from meningeal trigeminal nerve endings in the mechanism of migraine, blockade of neuropeptide release by anti-migraine drugs, and activation and sensitisation of trigeminal afferents by meningeal inflammatory stimuli and upstream role of intense brain activity. The study of neuropeptides provided the first evidence that antisera directed against calcitonin gene-related peptide (CGRP) and substance P could neutralise their actions. Successful therapeutic strategies using humanised monoclonal antibodies directed against CGRP and its receptor followed from these findings. Nowadays, 40 years after the initial proposal, the trigeminovascular system is widely accepted as having a fundamental role in this highly complex neurological disorder and provides a road map for future migraine therapies.
Collapse
Affiliation(s)
- Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jakob Møller Hansen
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thien Phu Do
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Agustin Melo-Carrillo
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Rami Burstein
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Michael A Moskowitz
- Department of Neurology and Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
10
|
Hargreaves R, Olesen J. Calcitonin Gene-Related Peptide Modulators - The History and Renaissance of a New Migraine Drug Class. Headache 2019; 59:951-970. [PMID: 31020659 DOI: 10.1111/head.13510] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2019] [Indexed: 01/31/2023]
Abstract
Several lines of evidence pointed to an important role for CGRP in migraine. These included the anatomic colocalization of CGRP and its receptor in sensory fibers innervating pain-producing meningeal blood vessels, its release by trigeminal stimulation, the observation of elevated CGRP in the cranial circulation during migraine with normalization concomitant with headache relief by sumatriptan, and translational studies with intravenous (IV) CGRP that evoked migraine only in migraineurs. The development of small molecule CGRP receptor antagonists (CGRP-RAs) that showed clinical antimigraine efficacy acutely and prophylactically in randomized placebo-controlled clinical trials subsequently gave definitive pharmacological proof of the importance of CGRP in migraine. More recently, CGRP target engagement imaging studies using a CGRP receptor PET ligand [11 C]MK-4232 demonstrated that there was no brain CGRP receptor occupancy at clinically effective antimigraine doses of telcagepant, a prototypic CGRP-RA. Taken together, these data indicated that (1) the therapeutic site of action of the CGRP-RAs was peripheral not central; (2) that IV CGRP had most likely evoked migraine through an action at sites outside the blood-brain barrier; and (3) that migraine pain was therefore, at least in part, peripheral in origin. The evolution of CGRP migraine science gave impetus to the development of peripherally acting drugs that could modulate CGRP chronically to prevent frequent episodic and chronic migraine. Large molecule biologic antibody (mAb) approaches that are given subcutaneously to neutralize circulating CGRP peptide (fremanezumab, galcanezumab) or block CGRP receptors (erenumab) have shown consistent efficacy and tolerability in multicenter migraine prevention trials and are now approved for clinical use. Eptinezumab, a CGRP neutralizing antibody given IV, shows promise in late stage clinical development. Recently, orally administered next-generation small molecule CGRP-RAs have been shown to have safety and efficacy in acute treatment (ubrogepant and rimegepant) and prevention (atogepant) of migraine, giving additional CGRP-based therapeutic options for migraine patients.
Collapse
Affiliation(s)
- Richard Hargreaves
- Center for Pain and the Brain, Harvard Medical School and Department of Anesthesia, Boston Children's Hospital, Boston, MA, USA
| | - Jes Olesen
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Glostrup, Denmark
| |
Collapse
|
11
|
Kozłowska A, Mikołajczyk A, Adamiak Z, Majewski M. Distribution and chemical coding of sensory neurons innervating the skin of the porcine hindlimb. Neuropeptides 2017; 61:1-14. [PMID: 27866657 DOI: 10.1016/j.npep.2016.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 10/17/2016] [Accepted: 10/24/2016] [Indexed: 11/18/2022]
Abstract
The aim of the present study was to establish the origin and chemical phenotyping of neurons involved in skin innervation of the porcine hind leg. The dorsal root ganglia (DRGs) of the lumbar (L4-L6) and sacral (S1-S3) spinal nerves were visualized using the fluorescent tracer Fast Blue (FB). The morphometric analysis of FB-positive (FB+)neurons showed that in the L4, L5, S1 and S2 DRGs, the small-sized perikarya constituted the major population, whereas in the L6 and S3 DRGs the medium-sized cells made up the major population. In all these ganglia, large-sized FB+ perikarya constituted only a small percentage of all FB+ neurons. Immunohistochemistry revealed that small- and medium-sized FB+ perikarya contained sensory markers such as: substance P (SP), calcitonin gene related peptide (CGRP) and galanin (GAL); as well as various other factors such as somatostatin (SOM), calbindin-D28k (CB), pituitary adenylate cyclase-activating polypeptide (PACAP) and neuronal nitric oxide synthase (nNOS). Meanwhile large-sized FB+ perikarya usually expressed SP, CGRP or PACAP. In the lumbar DRGs, some large cells also contained SOM and CB. Double-labeling immunohistochemistry showed that SP-positive neurons co-expressed CGRP, GAL or PACAP; while PACAP-positive cells co-expressed GAL or nNOS. Neurons stained for SOM were also immunoreactive for CB or GAL, while neurons stained for nNOS were also immunoreactive for GAL. In conclusion, the present data has indicated that the distribution and chemical phenotyping of the porcine skin-projecting neurons are different within DRGs of the lumbar (forming a femoral nerve) and sacral (forming a sciatic nerve) spinal nerves.
Collapse
Affiliation(s)
- Anna Kozłowska
- Department of Human Physiology, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Poland.
| | - Anita Mikołajczyk
- Department of Public Health, Epidemiology and Microbiology, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Poland
| | - Zbigniew Adamiak
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Poland
| | - Mariusz Majewski
- Department of Human Physiology, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Poland
| |
Collapse
|
12
|
Berczi I, Rotondo F, Kovacs K. Neuropeptides in Immunoregulation. INSIGHTS TO NEUROIMMUNE BIOLOGY 2016:133-181. [DOI: 10.1016/b978-0-12-801770-8.00007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Park JJ, Chon NR, Lee YJ, Park H. The Effects of an Extract of Atractylodes Japonica Rhizome, SKI3246 on Gastrointestinal Motility in Guinea Pigs. J Neurogastroenterol Motil 2015; 21:352-60. [PMID: 26130631 PMCID: PMC4496911 DOI: 10.5056/jnm14112] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 03/14/2015] [Accepted: 03/15/2015] [Indexed: 11/20/2022] Open
Abstract
Background/Aims There are limited therapeutic options available for irritable bowel syndrome with diarrhea (IBS-D). We tested the effects of Atractylodes japonica rhizome, a perennial plant native to North Asia, on both upper and lower gastrointestinal (GI) motility in guinea pigs. Methods The extract of A. japonica rhizome was administered orally at different doses to test its effects on upper GI motility as determined from charcoal transit in native guinea pigs and in guinea pigs pretreated with thyrotropin-releasing hormone or mustard oil. Regarding its effect on lower GI motility, the removed guinea pig colon was suspended in a chamber containing Krebs-Henseleit solution and the transit time of artificial feces was measured with various dilutions of the extract. As for in vivo assay, weight and number of fecal pellets expelled were determined under the same drug preparation used in upper GI motility experiment. Results The extract of A. japonica rhizome had no significant effect on upper GI motility in either normal or altered physiological states. However, the extract increased colonic transit time in the in vitro model. In the fecal expulsion study, the cumulative weight and number of pellets did not differ significantly between the control group and groups treated with the extracts. In the animals pretreated in vivo with thyrotropin-releasing hormone, however, the weight and number of fecal pellets were significantly decreased in animals treated with 300 mg/kg and 600 mg/kg doses of extract. Conclusions Our findings suggest that the extract of A. japonica rhizome can be a potential agent for IBS-D.
Collapse
Affiliation(s)
- Jae Jun Park
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Nu Ri Chon
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ju Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojin Park
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Ushio N, Dai Y, Wang S, Fukuoka T, Noguchi K. Transient receptor potential channel A1 involved in calcitonin gene-related peptide release in neurons. Neural Regen Res 2014; 8:3013-9. [PMID: 25206621 PMCID: PMC4146211 DOI: 10.3969/j.issn.1673-5374.2013.32.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 09/12/2013] [Indexed: 01/25/2023] Open
Abstract
Transient receptor potential channel A1 is one of the important transducers of noxious stimuli in the primary afferents, which may contribute to generation of neurogenic inflammation and hyperalgesia. The present study was designed to investigate if activation of transient receptor potential channel A1 may induce calcitonin gene-related peptide release from the primary afferent neurons. We found that application of allyl isothiocyanate, a transient receptor potential channel A1 activator, caused calcitonin gene-related peptide release from the cultured rat dorsal root ganglion neurons. Knockdown of transient receptor potential channel A1 with an antisense oligodeoxynucleotide prevented calcitonin gene-related peptide release by allyl isothiocyanate application in cultured dorsal root ganglion neurons. Thus, we concluded that transient receptor potential channel A1 activation caused calcitonin gene-related peptide release in sensory neurons.
Collapse
Affiliation(s)
- Nobumasa Ushio
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Yi Dai
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan ; Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo 650-8530, Japan ; Traditional Medicine Research Center, Chinese Medicine Confucius Institute at Hyogo College of Medicine, Kobe, Hyogo 650-8530, Japan
| | - Shenglan Wang
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan ; Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences, Kobe, Hyogo 650-8530, Japan ; Traditional Medicine Research Center, Chinese Medicine Confucius Institute at Hyogo College of Medicine, Kobe, Hyogo 650-8530, Japan
| | - Tetsuo Fukuoka
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Koichi Noguchi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
15
|
Neurobiological mechanisms of pelvic pain. BIOMED RESEARCH INTERNATIONAL 2014; 2014:903848. [PMID: 25110704 PMCID: PMC4119661 DOI: 10.1155/2014/903848] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/03/2014] [Indexed: 11/29/2022]
Abstract
Pelvic pain is a common condition which significantly deteriorates health-related quality of life. The most commonly identified causes of pain in the pelvic region are gynaecologic, urologic, gastrointestinal, neurological, and musculoskeletal. However, in up to 33% of patients the source of this symptom is not identified, frustrating both patients and health-care professionals. Pelvic pain may involve both the somatic and visceral systems, making the differential diagnosing challenging. This paper aimed to review the mechanisms involved in pelvic pain perception by analyzing the neural plasticity and molecules which are involved in these complex circuits.
Collapse
|
16
|
Bodkin JV, Thakore P, Aubdool AA, Liang L, Fernandes ES, Nandi M, Spina D, Clark JE, Aaronson PI, Shattock MJ, Brain SD. Investigating the potential role of TRPA1 in locomotion and cardiovascular control during hypertension. Pharmacol Res Perspect 2014; 2:e00052. [PMID: 25505598 PMCID: PMC4186440 DOI: 10.1002/prp2.52] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 12/23/2022] Open
Abstract
Radiotelemetry was used to investigate the in vivo cardiovascular and activity phenotype of both TRPA1 (transient receptor potential ankyrin 1) wild-type (WT) and TRPA1 knockout (KO) mice. After baseline recording, experimental hypertension was induced using angiotensin II infusion (1.1 mg(-1) kg(-1) a day, for 14 days). TRPA1 WT and KO mice showed similar morphological and functional cardiovascular parameters, including similar basal blood pressure (BP), heart rate, size, and function. Similar hypertension was also displayed in response to angiotensin II (156 ± 7 and 165 ± 11 mmHg, systolic BP ± SEM, n = 5-6). TRPA1 KO mice showed increased hypertensive hypertrophy (heart weight:tibia length: 7.3 ± 1.6 mg mm(-1) vs. 8.8 ± 1.7 mg mm(-1)) and presented with blunted interleukin 6 (IL-6) production compared with hypertensive WT mice (151 ± 24 vs. 89 ± 16 pg mL(-1)). TRPA1 expression in dorsal root ganglion (DRG) neurones was upregulated during hypertension (163% of baseline expression). Investigations utilizing the TRPA1 agonist cinnamaldehyde (CA) on mesenteric arterioles isolated from näive mice suggested a lack of TRPA1-dependent vasoreactivity in this vascular bed; a site with notable ability to alter total peripheral resistance. However, mesenteric arterioles isolated from TRPA1 KO hypertensive mice displayed significantly reduced ability to relax in response to nitric oxide (NO) (P < 0.05). Unexpectedly, naïve TRPA1 KO mice also displayed physical hyperactivity traits at baseline, which was exacerbated during hypertension. In conclusion, our study provides a novel cardiovascular characterization of TRPA1 KO mice in a model of hypertension. Results suggest that TRPA1 has a limited role in global cardiovascular control, but we demonstrate an unexpected capacity for TRPA1 to regulate physical activity.
Collapse
Affiliation(s)
- Jennifer V Bodkin
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K
| | - Pratish Thakore
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K ; Pharmaceutical Sciences Division, School of Biomedical Sciences, King's College London London, SE1 9NH, U.K
| | - Aisah A Aubdool
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K
| | - Lihuan Liang
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K
| | - Elizabeth S Fernandes
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K ; Programa de Pós-Graduação em Biologia Parasitária, Universidade Ceuma São Luís, Brazil
| | - Manasi Nandi
- Pharmaceutical Sciences Division, School of Biomedical Sciences, King's College London London, SE1 9NH, U.K
| | - Domenico Spina
- Pharmaceutical Sciences Division, School of Biomedical Sciences, King's College London London, SE1 9NH, U.K
| | - James E Clark
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K
| | - Philip I Aaronson
- Asthma, Allergy and Lung Biology Division, School of Medicine, King's College London London, SE1 1UL, U.K
| | - Michael J Shattock
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K
| | - Susan D Brain
- Cardiovascular Division, BHF Centre of Excellence and Centre of Integrative Biomedicine, School of Medicine, King's College London London, SE1 9NH, U.K
| |
Collapse
|
17
|
Innervation mapping of the hind paw of the rat using Evans Blue extravasation, Optical Surface Mapping and CASAM. J Neurosci Methods 2014; 229:15-27. [DOI: 10.1016/j.jneumeth.2014.03.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 11/20/2022]
|
18
|
Early demyelination of primary A-fibers induces a rapid-onset of neuropathic pain in rat. Neuroscience 2011; 200:186-98. [PMID: 22061425 DOI: 10.1016/j.neuroscience.2011.10.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 10/18/2011] [Accepted: 10/20/2011] [Indexed: 12/23/2022]
Abstract
Some types of peripheral neuropathic pain are associated with damage to myelin rather than to axons of primary sensory neurons. It is extremely important to develop selective demyelination animal models for understanding neuropathic pain caused by demyelination. We induced a rapid-onset and reversible demyelination of peripheral A-fibers and neuropathic pain behaviors in adult rats by a single injection of cobra venom into the sciatic nerve. The relation between A-fiber demyelination and the abnormal pain behaviors was investigated using this model. Microfilament recordings revealed that cobra venom selectively blocked A-fibers, but not C-fibers. Selective blockade of A-fibers may result from A-fiber demyelination at the site of venom injection as demonstrated by microscope examination. The axons of the demyelinated A-fibers appeared to be otherwise normal. Neuropathic pain behaviors appeared almost immediately after venom injection and lasted about 3 weeks. Electrophysiological studies indicated that venom injection induced loss of conduction in A-fibers, increased sensitivity of C-polymodal nociceptors to innocuous stimuli, and triggered spontaneous activity from both peripheral and central terminals of C-fiber nociceptors. Neurogenic inflammatory responses were also observed in the affected skin via Evan's Blue extravasation experiments. Both antidromic C-fiber spontaneous activity and neurogenic inflammation were substantially decreased by continuous A-fiber threshold electric stimuli applied proximally to the venom injection site. The data suggest that normal activity of peripheral A-fibers may produce inhibitory modulation of C-fiber polymodal nociceptors. Removal of inhibition to C-fiber polymodal nociceptors following demyelination of A-fibers may result in pain and neurogenic inflammation in the affected receptive field.
Collapse
|
19
|
Vajdy M. Immunomodulatory properties of vitamins, flavonoids and plant oils and their potential as vaccine adjuvants and delivery systems. Expert Opin Biol Ther 2011; 11:1501-13. [PMID: 21955085 DOI: 10.1517/14712598.2011.623695] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION During the past century, vaccinologists have attempted to mimic pathogens in their immune-enhancing capacity. This led to the development of life-saving vaccines based on live attenuated viruses, bacteria and toxoids. Hence, intense research in vaccine adjuvant discovery has focused on toll like receptors, mutant toxins and viral and bacterial vectors. Nutritive components such as vitamins and select polyphenols also possess immunomodulating properties without the potential toxic and adverse side effects of agents that mimic danger signals. AREAS COVERED This review pertains to immunomodulatory properties of nutritive components, that is vitamins A, C, D, E, flavonoids and plant oils, as potential vaccine adjuvants and delivery systems, covering Pubmed publication searches from 1980 through 2011. EXPERT OPINION This relatively unexplored field of the potential of nutritive components as vaccine adjuvants holds great promise to promote the development of effective and above all safe vaccines. Hence the future focus should be placed on enhancing their efficacy, mainly through novel approaches in designing structural derivatives, formulations, delivery systems and routes of administration. As safety has been the major issue in development of novel vaccines, this new approach will probably result in new discoveries in designing safe and effective vaccines.
Collapse
Affiliation(s)
- Michael Vajdy
- EpitoGenesis, Inc., 1810 North Broadway, Walnut Creek, CA 94596, USA.
| |
Collapse
|
20
|
Umukoro S, Olugbemide AS. Antinociceptive effects of methyl jasmonate in experimental animals. J Nat Med 2011; 65:466-70. [DOI: 10.1007/s11418-011-0520-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 02/06/2011] [Indexed: 10/18/2022]
|
21
|
Yu M, Vajdy M. A novel retinoic acid, catechin hydrate and mustard oil-based emulsion for enhanced cytokine and antibody responses against multiple strains of HIV-1 following mucosal and systemic vaccinations. Vaccine 2011; 29:2429-36. [PMID: 21272602 DOI: 10.1016/j.vaccine.2011.01.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 12/10/2010] [Accepted: 01/12/2011] [Indexed: 01/05/2023]
Abstract
Non-replicating protein- or DNA-based antigens generally require immune-enhancing adjuvants and delivery systems. It has been particularly difficult to raise antibodies against gp120 of HIV-1, which constitutes an important approach in HIV vaccine design. While almost all effort in adjuvant research has focused on mimicking the pathogens and the danger signals they engender in the host, relatively little effort has been spent on nutritive approaches. In this study, a new nutritive immune-enhancing delivery system (NIDS) composed of vitamin A, a polyphenol-flavonoid, catechin hydrate, and mustard oil was tested for its adjuvant effect in immune responses against the gp120 protein of HIV-1(CN54). Following a combination of two mucosal and two systemic vaccinations of mice, we found significant enhancement of both local and systemic antibodies as well as cytokine responses. These data have important implications for vaccine and adjuvant design against HIV-1 and other pathogens.
Collapse
Affiliation(s)
- Mingke Yu
- EpitoGenesis, Inc. 1810 North Broadway, Walnut Creek, CA 94596, USA
| | | |
Collapse
|
22
|
|
23
|
Wu ZM, Chen YF, Qiu PN, Ling SC. Correlation between the distribution of SP and CGRP immunopositive neurons in dorsal root ganglia and the afferent sensation of preputial frenulum. Anat Rec (Hoboken) 2010; 294:479-86. [PMID: 21337713 DOI: 10.1002/ar.21327] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 10/16/2010] [Accepted: 11/09/2010] [Indexed: 11/08/2022]
Abstract
The aim of this study was to explore the distribution of substance P (SP) and calcitonin gene-related peptide (CGRP) immunoreactive nerve terminals in the penis prepuce and the preputial frenulum. The possible correlation between SP- and CGRP-immunopositive neurons in dorsal root ganglia (DRG) and the afferent sensation of the penile preputial frenulum is also discussed. Immunohistochemistry showed SP- and CGRP-positive nerve terminals in the epidermal basal layer of the prepuce and frenulum in adult human males. The majority of the nerve terminals presented as bundles of different lengths and a few as enlarged nodosities. The density of SP- and CGRP-immunopositive nerve terminals in the preputial frenulum was significantly higher than those in the penis prepuce (P<0.01). Fluoro-Gold (FG) retrograde tracing method was used to trace the origin of nerve terminals in Sprague-Dawley rats. SP and CGRP immunofluorescence labeling was employed to detect the distribution of SP- and CGRP-immunoreactive neurons in DRG. FG retro-labeled neurons were localized in L(6) -DRG and S(1) -DRG. All the FG/SP and FG/CGRP double-labeled neurons were medium or small-sized. One-third of the FG-labeled neurons were SP-immunoreactive, and a half of them CGRP-immunoreactive in L(6) -DRG and S(1) -DRG, respectively. The FG/SP/CGRP-labeled neurons accounted for one fifth of the FG retro-labeled neurons. Taken together, these data suggest that the SP- and CGRP-immunopositive nerve fibers may participate in the transmission of afferent sensation in the preputial frenulum.
Collapse
Affiliation(s)
- Zhong-Min Wu
- Department of Anatomy, School of Medicine of Zhejiang University, Hangzhou, China
| | | | | | | |
Collapse
|
24
|
Hagains CE, Trevino LA, He JW, Liu H, Peng YB. Contributions of dorsal root reflex and axonal reflex to formalin-induced inflammation. Brain Res 2010; 1359:90-7. [PMID: 20816764 DOI: 10.1016/j.brainres.2010.08.097] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 08/25/2010] [Accepted: 08/28/2010] [Indexed: 02/06/2023]
Abstract
The dorsal root reflex (DRR) and the axonal reflex (AR) are antidromic activities in primary afferents and are involved in neurogenic inflammation. DRRs and/or ARs lead to release of neuropeptides calcitonin gene-related peptide (CGRP) and substance P (SP). CGRP causes blood vessels to dilate leading to an increase in blood perfusion, whereas SP causes plasma extravasation, leading to edema. Both DRR and AR can be evoked by noxious stimuli. The goal of this study was to determine the role of DRR and AR in neurogenic inflammation by examining the blood perfusion (BP) change in hindpaws in response to formalin injection (an acute inflammatory agent). Laser Doppler images were collected simultaneously in both hindpaws in anesthetized rats to determine the level of BP. Local lidocaine was applied to the left sciatic nerve to block both orthodromic signals and antidromic DRRs without affecting ARs. All rats then received a subcutaneous formalin injection to the left hindpaw. Our results showed that (1) the mean BP of the left paw increased significantly following formalin injection, with or without lidocaine; (2) application of lidocaine in the left sciatic nerve alone significantly increased BP ipsilaterally; (3) formalin injection following lidocaine application significantly increased BP more than the group without lidocaine; and (4) there was delayed significant BP increase in the right (contralateral) hindpaw following formalin injection with or without lidocaine. It is concluded that ARs play a more important role than DRRs in formalin-induced neurogenic inflammation.
Collapse
|
25
|
Effects of a bupivacaine nerve block on the axonal transport of Tumor Necrosis Factor-alpha (TNF-alpha) in a rat model of carrageenan-induced inflammation. Brain Behav Immun 2010; 24:652-9. [PMID: 20144702 DOI: 10.1016/j.bbi.2010.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 01/27/2010] [Accepted: 01/31/2010] [Indexed: 11/23/2022] Open
Abstract
Many pro-inflammatory cytokines are involved in the process of inflammatory pain. Bi directional axonal transport of Tumor Necrosis Factor-alpha (TNF-alpha) occurs in case of neuropathic pain induced by nerve ligation. We used an in vivo preparation with injection of carrageenan and fluorescent TNF-alpha in the territory of the saphenous nerve of rats to study this transport. We have shown that retrograde transport of TNF-alpha occurs after an inflammatory insult caused by the injection of carrageenan. This transport was likely mediated by the TNF receptor 1. A nerve block with bupivacaine totally abolishes the expression of the receptor in the dorsal root ganglion and the retrograde transport of TNF-alpha. In addition, bupivacaine at low concentrations (1-10 microM) was able to stop the axonal transport ex vivo. Tetrodotoxin was less efficacious for inhibiting the TNF-alpha transport and the rise in receptor expression and for inhibiting the axonal transport ex vivo. This may partly explain the efficacy of nerve blocks with bupivacaine to decrease the neurogenic inflammation and in a lower extent the long-term inhibition of hyperalgesic phenomenon observed in animals and in humans.
Collapse
|
26
|
Bonjardim LR, da Silva AP, Gameiro GH, Tambeli CH, Ferraz de Arruda Veiga MC. Nociceptive behavior induced by mustard oil injection into the temporomandibular joint is blocked by a peripheral non-opioid analgesic and a central opioid analgesic. Pharmacol Biochem Behav 2008; 91:321-6. [PMID: 18755210 DOI: 10.1016/j.pbb.2008.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2008] [Revised: 07/12/2008] [Accepted: 08/01/2008] [Indexed: 10/21/2022]
Abstract
The aim of this study was to improve the mustard oil (MO) induced temporomandibular joint (TMJ) nociception model and to investigate the potential analgesic activity of systemic dipyrone and tramadol on the nociceptive behavioral responses induced by injection of low concentrations of the MO into the rat TMJ region. TMJ injection of 2.5% MO produced a significant nociceptive behavior expressed by head flinching and orofacial rubbing. This activity was related to the MO injection since mineral oil (vehicle) did not elicit response. Local application of the lidocaine N-ethyl bromide quaternary salt, QX-314 (2%) and systemic administration of morphine (4 mg/kg) significantly reduced the MO-induced nociceptive responses, validating the nociceptive character of the behaviors. The pretreatment with systemic dipyrone (19, 57 or 95 mg/kg) as well as tramadol (5, 7.5 or 10 mg/kg) was effective in decreasing the nociceptive behavioral responses induced by the injection of MO into the rat TMJ. In conclusion, TMJ injection of low concentrations of MO in rats produces well defined and quantifiable nociceptive behaviors constituting a reliable behavioral model for studying TMJ pain mechanisms and testing analgesic drugs. The results also suggest that dipyrone and tramadol could be effective analgesic options in the management of TMJ pain.
Collapse
|
27
|
Carmichael NME, Dostrovsky JO, Charlton MP. Enhanced vascular permeability in rat skin induced by sensory nerve stimulation: evaluation of the time course and appropriate stimulation parameters. Neuroscience 2008; 153:832-41. [PMID: 18420352 DOI: 10.1016/j.neuroscience.2008.02.068] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 02/25/2008] [Accepted: 02/29/2008] [Indexed: 11/29/2022]
Abstract
Activation of nociceptors causes them to secrete neuropeptides. The binding of these peptides to receptors on blood vessels causes vasodilation and increased vascular permeability that allows loss of proteins and fluid (plasma extravasation, PE); this contributes to inflammation. This study defines the relationship between electrical activation of nociceptors and PE and evaluates the time course of this response in the skin of rats. We measured the time course and extent of PE by digital imaging of changes in skin reflectance caused by leakage of Evans Blue (EB) dye infused in the circulatory system before stimulation. Stimulation of the exclusively sensory saphenous nerve caused the skin to become dark blue within 2 min due to accumulation of EB. While PE is usually measured after 5-15 min of electrical stimulation, we found that stimulation for only 1 min at 4 Hz produced maximum PE. This response was dependent on the number of electrical stimuli at least for 4 Hz and 8 Hz stimulation rates. Since accumulation of EB in the skin is only slowly reversible, to determine the duration of enhanced vascular permeability we administered EB at various times after electrical stimulation of the saphenous nerve. PE was only observed when EB was infused within 5 min of electrical stimulation but could still be observed 50 min after capsaicin (1%, 25 microl) injection into the hind paw. These findings indicate that enhanced vascular permeability evoked by electrical stimulation persists only briefly after release of neuropeptides from nociceptors in the skin. Therefore, treatment of inflammation by blockade of neuropeptide release and receptors may be more effective than treatments aimed at epithelial gaps. We propose, in models of stimulation-induced inflammation, the use of a short stimulus train.
Collapse
Affiliation(s)
- N M E Carmichael
- University of Toronto, Department of Physiology, Toronto, Ontario, Canada.
| | | | | |
Collapse
|
28
|
Lin Q, Li D, Xu X, Zou X, Fang L. Roles of TRPV1 and neuropeptidergic receptors in dorsal root reflex-mediated neurogenic inflammation induced by intradermal injection of capsaicin. Mol Pain 2007; 3:30. [PMID: 17961222 PMCID: PMC2174436 DOI: 10.1186/1744-8069-3-30] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2007] [Accepted: 10/25/2007] [Indexed: 05/25/2023] Open
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 μg. 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.
Collapse
Affiliation(s)
- Qing Lin
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, Texas 77555-1069, USA.
| | | | | | | | | |
Collapse
|
29
|
Malykhina AP. Neural mechanisms of pelvic organ cross-sensitization. Neuroscience 2007; 149:660-72. [PMID: 17920206 DOI: 10.1016/j.neuroscience.2007.07.053] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 07/05/2007] [Accepted: 07/12/2007] [Indexed: 12/12/2022]
Abstract
Clinical observations of viscerovisceral referred pain in patients with gastrointestinal and genitourinary disorders suggest an overlap of neurohumoral mechanisms underlying both bowel and urinary bladder dysfunctions. Close proximity of visceral organs within the abdominal cavity complicates identification of the exact source of chronic pelvic pain, where it originates, and how it relocates with time. Cross-sensitization among pelvic structures may contribute to chronic pelvic pain of unknown etiology and involves convergent neural pathways of noxious stimulus transmission from two or more organs. Convergence of sensory information from discrete pelvic structures occurs at different levels of nervous system hierarchy including dorsal root ganglia, the spinal cord and the brain. The cell bodies of sensory neurons projecting to the colon, urinary bladder and male/female reproductive organs express a wide range of membrane receptors and synthesize many neurotransmitters and regulatory peptides. These substances are released from nerve terminals following enhanced neuronal excitability and may lead to the occurrence of neurogenic inflammation in the pelvis. Multiple factors including inflammation, nerve injury, ischemia, peripheral hyperalgesia, metabolic disorders and other pathological conditions dramatically alter the function of directly affected pelvic structures as well as organs located next to a damaged domain. Defining precise mechanisms of viscerovisceral cross-sensitization would have implications for the development of effective pharmacological therapies for the treatment of functional disorders with chronic pelvic pain such as irritable bowel syndrome and painful bladder syndrome. The complexity of overlapping neural pathways and possible mechanisms underlying pelvic organ crosstalk are analyzed in this review at both systemic and cellular levels.
Collapse
Affiliation(s)
- A P Malykhina
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Boulevard, Oklahoma City, OK 73104, USA.
| |
Collapse
|
30
|
Juhl L, Edvinsson L, Olesen J, Jansen-Olesen I. Effect of two novel CGRP-binding compounds in a closed cranial window rat model. Eur J Pharmacol 2007; 567:117-24. [PMID: 17477918 DOI: 10.1016/j.ejphar.2007.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 03/28/2007] [Accepted: 04/01/2007] [Indexed: 11/19/2022]
Abstract
We investigated the in vivo effects of two novel calcitonin gene-related peptide (CGRP) binding molecules in the genuine closed cranial window model in the rat. The RNA-Spiegelmer (NOX-C89) and the monoclonal CGRP antibody are CGRP scavengers and might be used as an alternative to CGRP-receptor antagonists in the treatment of migraine. Rats were anaesthetized and a closed cranial window established. Changes in dural and pial artery diameter and mean arterial blood pressure were measured simultaneously. Infusion of the RNA-Spiegelmer or the CGRP antibody alone had no effect on the arteries or the mean arterial blood pressure. We then used a bolus of 0.3 microg/kg CGRP (n=6) or electrical stimulation (25 V, 5 Hz, 1 ms pulse width and of 10 s of duration) (n=6) to induce dilatation of dural and pial arteries (mediated via CGRP-receptors). Pre-treatment with the RNA-Spiegelmer inhibited CGRP-induced vasodilatation of the dural artery (from 38+/-17% to 7+/-3%) and the pial artery (from 14+/-1% to 3+/-2%) (P<0.05). The RNA-Spiegelmer, however, did not significantly inhibit dilatation induced by electrical stimulation (P>0.05). The CGRP antibody caused a significant reduction of the dural artery diameter caused by intravenous CGRP-infusion (from 23+/-5% to 12+/-3%) (P<0.05), but did not inhibit dilatation caused by electrical stimulation (P>0.05). In conclusion, the CGRP scavengers effectively inhibited the effect of circulating CGRP but do not modify the effect of electrical stimulation and the consequent liberation of CGRP from perivascular sensory nerve fibres.
Collapse
Affiliation(s)
- Louise Juhl
- Department of Neurology, Glostrup Research Institute, Glostrup Hospital, University of Copenhagen, DK-2600, Glostrup, Denmark.
| | | | | | | |
Collapse
|
31
|
Bermúdez R, Vigliano F, Quiroga MI, Nieto JM, Bosi G, Domeneghini C. Immunohistochemical study on the neuroendocrine system of the digestive tract of turbot, Scophthalmus maximus (L.), infected by Enteromyxum scophthalmi (Myxozoa). FISH & SHELLFISH IMMUNOLOGY 2007; 22:252-63. [PMID: 16844386 DOI: 10.1016/j.fsi.2006.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Revised: 05/23/2006] [Accepted: 05/30/2006] [Indexed: 05/10/2023]
Abstract
In recent years a new parasite, causing severe losses, has been detected in farmed turbot, Scophthalmus maximus (L.), in Northwestern Spain. Dead fish showed emaciation and cachexia caused by severe necrotizing enteritis, which affected all areas of the digestive tract. The parasite was classified as a myxosporean and named Enteromyxum scophthalmi. This study was designed to assess the response of the turbot neuroendocrine system against E. scophthalmi infection. Immunohistochemical tests were applied to sections of the gastrointestinal tract of uninfected and E. scophthalmi-infected turbot, and the presence of cholecystokinin (CCK-8), serotonin (5-HT), substance P (SP), calcitonin gene-related peptide (CGRP) and vasoactive intestinal peptide (VIP) were documented. A higher abundance of both endocrine epithelial cells (ECs) and nerve cell bodies and fibres for CCK-8, 5-HT and SP were recorded in the gastrointestinal tract of infected turbot, whereas VIP-like substance decreased. The results indicate that E. scophthalmi infection in turbot induced changes in the neuroendocrine system, which may cause alterations in gut motility, electrolyte and fluid secretion, and vascular and immune functions.
Collapse
Affiliation(s)
- R Bermúdez
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, University of Santiago de Compostela, 27002 Lugo, Spain.
| | | | | | | | | | | |
Collapse
|
32
|
Valencia-de Ita S, Lawand NB, Lin Q, Castañeda-Hernandez G, Willis WD. Role of the Na+-K+-2Cl- cotransporter in the development of capsaicin-induced neurogenic inflammation. J Neurophysiol 2006; 95:3553-61. [PMID: 16709721 DOI: 10.1152/jn.01091.2005] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent behavioral and electrophysiological studies have attributed an important role to dorsal root reflexes (DRRs) in the initiation and development of neurogenic inflammation produced by intradermal capsaicin (CAP). The DRRs can occur in peptidergic fibers, resulting in peripheral release of neuromediators that produce vasodilation, plasma extravasation and subsequently hyperalgesia and allodynia. In this study, we have evaluated the effect of spinal administration of bumetanide (a blocker of the Na+-K+-2Cl- cotransporter, NKCC) on DRR activity, changes in cutaneous blood flow (vasodilation), hindpaw edema, mechanical allodynia, and hyperalgesia induced by intradermal injection of 1% CAP in Sprague-Dawley rats. Vasodilation was monitored using laser Doppler flowmetry, neurogenic edema was evaluated by measurements of hindpaw volume, and secondary mechanical allodynia and hyperalesia were tested using von Frey filaments (10 and 200 mN) applied to the plantar surface of the paw. Changes in the blood flow were blocked significantly by intrathecal bumetanide at 10 and 100 microM in both pre- and posttreatment studies. Spinal bumetanide at 10 and 100 microM blocked neurogenic edema when it was administered before CAP injection, but only bumetanide at 100 microM administered after CAP injection reduced the paw edema significantly. Furthermore, the administration of bumetanide onto the spinal cord reduced the increment in DRR activity produced by CAP. Finally, both secondary mechanical allodynia and hyperalesia were reduced by bumetanide at 1, 10, and 100 microM. Taken together these results suggest that NKCC is involved in the increases in DRR activity, neurogenic inflammation and hyperalgesia and allodynia induced by intradermal CAP.
Collapse
Affiliation(s)
- Sandra Valencia-de Ita
- Seccion Externa de Farmacologia, Centro de Investigacion y de Estudios Avanzados del Instituto Politecnico Nacional, Mexico, DF Mexico
| | | | | | | | | |
Collapse
|
33
|
Holzer P. Peptidergic sensory neurons in the control of vascular functions: mechanisms and significance in the cutaneous and splanchnic vascular beds. Rev Physiol Biochem Pharmacol 2005; 121:49-146. [PMID: 1485073 DOI: 10.1007/bfb0033194] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- P Holzer
- University of Graz, Department of Experimental and Clinical Pharmacology, Austria
| |
Collapse
|
34
|
Gonzalez HL, Carmichael N, Dostrovsky JO, Charlton MP. Evaluation of the Time Course of Plasma Extravasation in the Skin by Digital Image Analysis. THE JOURNAL OF PAIN 2005; 6:681-8. [PMID: 16202961 DOI: 10.1016/j.jpain.2005.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Revised: 06/07/2005] [Accepted: 06/24/2005] [Indexed: 11/24/2022]
Abstract
UNLABELLED Plasma extravasation (PE) can be triggered by neurotransmitters as part of a neuroinflammatory response. We present a technique based on video digital image processing that provides a simple, noninvasive, reliable, and quantitative method for measuring the time course and extent of PE in the skin. After intravenous infusion of Evans Blue dye, stimulation of the saphenous nerve caused the skin on the dorsomedial region of the hind paw to become dark blue. The change in reflectance of the skin was recorded with a monochrome video camera. Images were digitized and analyzed with inexpensive or public domain software. The change in pixel intensity was determined in a selected region. Stimulation at 4 Hz caused greater darkening of the skin than at 1 Hz, and this was confirmed with spectrophotometric measurements of Evans Blue content. The NK1 receptor antagonist CP-99, 994 blocked saphenous nerve and substance P-induced darkening of the skin. The results indicate that our measurement gives results similar to those obtained with classic methods that are widely accepted as an indication of PE. This simple and quick method reveals the extent, time course, and location of PE, is cheap to implement and easy to learn, and thus represents a useful and alternative tool for studies of PE and its modulation. PERSPECTIVE This article presents a simple technique with which to evaluate the time course and extent of plasma extravasation in the skin of animal models of neuroinflammation. The technique is well suited to answer questions about basic physiologic mechanisms of neuroinflammation and should also be useful in drug testing studies.
Collapse
|
35
|
Kimball ES, Palmer JM, D'Andrea MR, Hornby PJ, Wade PR. Acute colitis induction by oil of mustard results in later development of an IBS-like accelerated upper GI transit in mice. Am J Physiol Gastrointest Liver Physiol 2005; 288:G1266-73. [PMID: 15691868 DOI: 10.1152/ajpgi.00444.2004] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Oil of mustard (OM) is a potent neuronal activator that promotes allodynia and hyperalgesia within minutes of application. In this study, OM was used to induce an acute colitis. We also investigated whether intracolonic OM-induced inflammation alters gastrointestinal (GI) function over a longer time frame as a model of postinflammatory irritable bowel syndrome (PI-IBS). Mice given a single administration of 0.5% OM developed a severe colitis that peaked at day 3, was reduced at day 7, and was absent by day 14. At the peak response, there was body weight loss, colon shrinkage, thickening and weight increases, distension of the proximal colon, and diarrhea. Macroscopic inspection of the distal colon revealed a discontinuous pattern of inflammatory damage and occasional transmural ulceration. Histological examination showed loss of epithelium, an inflammatory infiltrate, destruction of mucosal architecture, edema, and loss of circular smooth muscle architecture. OM administration increased transit of a carmine dye bolus from 58% of the total length of the upper GI tract in untreated age-matched controls to as high as 74% when tested at day 28 post-OM. Mice in the latter group demonstrated a significantly more sensitive response to inhibition of upper GI transit by the mu-opioid receptor agonist loperamide compared with normal mice. OM induces a rapid, acute, and transient colitis and, in the longer term, functional changes in motility that are observed when there is no gross inflammation and thereby is a model of functional bowel disorders that mimic aspects of PI-IBS in humans.
Collapse
Affiliation(s)
- Edward S Kimball
- Enterology Research Team, Johnson and Johnson Pharmaceutical, Research and Development, Welsh and McKean Roads, PO Box 776, Spring House, PA, 19477-0776, USA.
| | | | | | | | | |
Collapse
|
36
|
Larauche M, Anton PM, Peiro G, Eutamène H, Buéno L, Fioramonti J. Role of capsaicin-sensitive afferent nerves in different models of gastric inflammation in rats. Auton Neurosci 2004; 110:89-97. [PMID: 15046732 DOI: 10.1016/j.autneu.2003.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2003] [Revised: 10/28/2003] [Accepted: 11/27/2003] [Indexed: 11/25/2022]
Abstract
Capsaicin-sensitive afferent nerves are described as being protective against gastric inflammation; their destruction leads to an exacerbation of inflammatory processes. However, these nerves have been shown to exert a pro-inflammatory action on stress-induced gastritis in rats. Our study aimed to investigate the role of capsaicin-sensitive afferent nerves in different experimental models of gastritis in rats. Functional ablation of sensory nerves was achieved by systemic capsaicin treatment (100 mg/kg). Gastritis was induced by mild (iodoacetamide, diquat, surgical duodeno-gastric reflux [DGR]) and strong (70% ethanol, indomethacin) inflammatory agents. Antagonists of the CGRP1 and NK1 receptors, hCGRP8-37 and SR140333, were administered in rats treated with iodoacetamide and ethanol. Macroscopic damage scores (MDS), myeloperoxidase (MPO) activity and malondialdehyde (MDA) concentration were evaluated after sacrifice. Macroscopic lesions appeared only in ethanol and indomethacin gastritis and were enhanced by capsaicin treatment. Gastric MPO activity was significantly increased by all agents compared to controls. Capsaicin treatment did not have any effect on MPO activity in indomethacin-treated rats or in rats submitted to surgery for duodeno-gastric reflux. However, it abolished the increase in MPO induced by iodoacetamide and diquat, and significantly enhanced that induced by ethanol. hCGRP8-37 and SR140333 abolished the increase in MPO activity and MDA concentration in iodoacetamide treated rats. In ethanol-treated rats, SR140333 diminished MPO activity. These results indicate that, depending upon the nature and duration of the experimental inflammation, capsaicin-sensitive afferent nerves may act differently to control gastric inflammatory processes, suggesting the involvement of a neurogenic component in some forms of gastric inflammation.
Collapse
Affiliation(s)
- Muriel Larauche
- Neuro-Gastroenterology and Nutrition Unit, INRA, 180 chemin de Tournefeuille, BP 3, 31931 Toulouse cedex 9, France
| | | | | | | | | | | |
Collapse
|
37
|
Jordt SE, Bautista DM, Chuang HH, McKemy DD, Zygmunt PM, Högestätt ED, Meng ID, Julius D. Mustard oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature 2004; 427:260-5. [PMID: 14712238 DOI: 10.1038/nature02282] [Citation(s) in RCA: 1463] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Accepted: 12/12/2003] [Indexed: 11/08/2022]
Abstract
Wasabi, horseradish and mustard owe their pungency to isothiocyanate compounds. Topical application of mustard oil (allyl isothiocyanate) to the skin activates underlying sensory nerve endings, thereby producing pain, inflammation and robust hypersensitivity to thermal and mechanical stimuli. Despite their widespread use in both the kitchen and the laboratory, the molecular mechanism through which isothiocyanates mediate their effects remains unknown. Here we show that mustard oil depolarizes a subpopulation of primary sensory neurons that are also activated by capsaicin, the pungent ingredient in chilli peppers, and by Delta(9)-tetrahydrocannabinol (THC), the psychoactive component of marijuana. Both allyl isothiocyanate and THC mediate their excitatory effects by activating ANKTM1, a member of the TRP ion channel family recently implicated in the detection of noxious cold. These findings identify a cellular and molecular target for the pungent action of mustard oils and support an emerging role for TRP channels as ionotropic cannabinoid receptors.
Collapse
Affiliation(s)
- Sven-Eric Jordt
- Department of Cellular and Molecular Pharmacology University of California, San Francisco, California 94143-2140, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Bánvölgyi A, Pozsgai G, Brain SD, Helyes ZS, Szolcsányi J, Ghosh M, Melegh B, Pintér E. Mustard oil induces a transient receptor potential vanilloid 1 receptor-independent neurogenic inflammation and a non-neurogenic cellular inflammatory component in mice. Neuroscience 2004; 125:449-59. [PMID: 15062987 DOI: 10.1016/j.neuroscience.2004.01.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2004] [Indexed: 10/26/2022]
Abstract
A neurogenic component has been suggested to play a pivotal role in a range of inflammatory/immune diseases. Mustard oil (allyl-isothiocyanate) has been used in studies of inflammation to mediate neurogenic vasodilatation and oedema in rodent skin. The aim of the present study was to analyse mustard oil-induced oedema and neutrophil accumulation in the mouse ear focussing on the roles of neurokinin 1 (NK(1)) and vanilloid (TRPV1) receptors using normal (BALB/c, C57BL/6) as well as NK(1) and TRPV1 receptor knockout mice. A single or double treatment of 1% mustard oil on the BALB/c mouse ear induced ear oedema with responses diminished by 6 h. However a 25-30% increase in ear thickness was maintained by the hourly reapplication of mustard oil. Desensitisation of sensory nerves with capsaicin, or the NK(1) receptor antagonist SR140333, inhibited oedema but only in the first 3 h. Neutrophil accumulation in response to mustard oil was inhibited neither by SR140333 nor capsaicin pre-treatment. An activating dose of capsaicin (2.5%) induced a large oedema in C57BL/6 wild-type mice that was minimal in TRPV1 receptor knockout mice. By comparison, mustard oil generated ear swelling was inhibited by SR140333 in wild-type and TRPV1 knockout mice. Repeated administration of mustard oil maintained 35% oedema in TRPV1 knockout animals and the lack of TRPV1 receptors did not alter the leukocyte accumulation. In contrast repeated treatment caused about 20% ear oedema in Sv129+C57BL/6 wild-type mice but the absence of NK(1) receptors significantly decreased the response. Neutrophil accumulation showed similar values in both groups. This study has revealed that mustard oil can act via both neurogenic and non-neurogenic mechanisms to mediate inflammation in the mouse ear. Importantly, the activation of the sensory nerves was still observed in TRPV1 knockout mice indicating that the neurogenic inflammatory component occurs via a TRPV1 receptor independent process.
Collapse
Affiliation(s)
- A Bánvölgyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Pécs, Szigeti u. 12, H-7624 Pécs, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Dux M, Sántha P, Jancsó G. Capsaicin-sensitive neurogenic sensory vasodilatation in the dura mater of the rat. J Physiol 2003; 552:859-67. [PMID: 12949222 PMCID: PMC2343470 DOI: 10.1113/jphysiol.2003.050633] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The neurogenic sensory vascular responses of the dura mater encephali are considered to contribute significantly to the mechanisms of meningeal nociception and headache. Although the fundamental role of capsaicin-sensitive afferent nerves in the development of the neurogenic inflammatory responses of a variety of tissues is well established, their participation in meningeal vascular reactions is unclear. In the present study, the effects of the topical application of capsaicin on the dural blood flow and on the morphology of the dural nerve fibres were examined in control and capsaicin-pretreated rats by means of laser Doppler flowmetry and electron microscopy, respectively. In the control rats, the dural application of capsaicin at concentrations of 50 and 100 nM induced significant increases in blood flow in the branches of the medial meningeal artery. This capsaicin-induced vasodilatation was abolished by capsazepine, a transient receptor potential vanilloid 1 (TRPV1) receptor antagonist, and by hCGRP8-37, a calcitonin gene-related peptide (CGRP) receptor antagonist. Administration of capsaicin at higher concentrations (1 and 10 microM) resulted in marked, dose-dependent decreases in dural blood flow. The capsaicin-induced vasodilatation was abolished, whereas vasoconstriction was augmented, by systemic pretreatment of the animals with capsaicin. Electron microscopy revealed degenerating unmyelinated axons in the dura mater after an acute exposure to capsaicin (10 microM), providing support for the existence and possible functional role of capsaicin-sensitive dural afferent nerves. The results indicate that capsaicin-induced vasodilatation in the rat dura mater is mediated by the release of CGRP from the sensory nerves, whereas the vasoconstrictor response may be attributed to a direct action of capsaicin on the vascular smooth muscle. The present study demonstrates for the first time that capsaicin-sensitive nociceptive afferent nerves contribute significantly to the dural vasodilatory responses and suggests an important role in meningeal nociception.
Collapse
Affiliation(s)
- Maria Dux
- Department of Physiology, University of Szeged, Dóm tér 10, H-6720 Szeged, Hungary.
| | | | | |
Collapse
|
40
|
Domoki F, Sántha P, Bari F, Jancsó G. Perineural capsaicin treatment attenuates reactive hyperaemia in the rat skin. Neurosci Lett 2003; 341:127-30. [PMID: 12686382 DOI: 10.1016/s0304-3940(03)00191-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The neural mechanisms of reactive cutaneous hyperaemia were studied by using a novel experimental approach: the simultaneous measurement of cutaneous blood flow by laser-Doppler flowmetry in adjacent innervated and chemodenervated skin regions of the rat hindpaw served by the same artery. Transient occlusion of the femoral artery (0.5-6 min) resulted in reactive hyperaemia that was greatly reduced in the chemodenervated region. After 3 min arterial occlusion, peak cutaneous blood flow was 109+/-13% vs. 53+/-7%* (% change from baseline, n=11, *P<0.05), and the total hyperaemic response was 110+/-21 vs. 52+/-12* (arbitrary perfusion units) in intact vs. chemodenervated skin regions, respectively. The findings provide clear evidence for the involvement of peptidergic capsaicin-sensitive afferent nerves in the mechanism of reactive cutaneous hyperaemia.
Collapse
Affiliation(s)
- Ferenc Domoki
- Department of Physiology, Faculty of Medicine, University of Szeged, Dóm tér 10, H-6720, Szeged, Hungary
| | | | | | | |
Collapse
|
41
|
Takahashi Y, Hirayama J, Nakajima Y. Segmental regulation pattern of body surface temperature in the rat hindlimb. Brain Res 2002; 947:100-9. [PMID: 12144858 DOI: 10.1016/s0006-8993(02)02915-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Body surface zones or 'thermatomes', whose temperature is regulated by a single spinal segment, were investigated by thermography in the rat hindlimb. First, the spatial relation between the dermatome delineated by dye extravasation and the corresponding thermatome was investigated in rats pretreated with intravenous application of Evans blue. Electrical stimulation of the spinal nerves and sympathetic trunk segments at L3 and L5 induced a distal dominant temperature decrease. In contrast, Evans blue extravasation appeared in the medial (in L3) and lateral (in L5) paw only by electrical stimulation of the spinal nerves. Second, thermatomes L1-L5 were determined in other rats. Electrical stimulation of the sympathetic trunk segments L1-L6 produced a temperature decrease in the abdomen, hindlimb, and tail. However, the hindlimb temperature was regulated mainly by L2-L5 levels, particularly by L4 and L5. The abdomen was regulated uniformly by L1-L6, and the tail by L6. It was demonstrated that thermatomes are manifested differently from the corresponding dermatomes in the rat hindlimb.
Collapse
Affiliation(s)
- Yuzuru Takahashi
- Departments of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.
| | | | | |
Collapse
|
42
|
Kauppila T, Kontinen VK, Wei H, Jyväsjärvi E, Pertovaara A. Cutaneous vascular responses evoked by noxious stimulation in rats with the spinal nerve ligation-induced model of neuropathy. Brain Res Bull 2002; 58:21-6. [PMID: 12121808 DOI: 10.1016/s0361-9230(02)00751-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Antidromic activation of nociceptive nerve fibres innervating the skin produces an axon reflex that involves extravasation and vasodilation of cutaneous blood vessels. We determined whether the axon reflex of the hindlimb skin is influenced by an experimental model of neuropathy induced by unilateral ligation of spinal nerves L(5) and L(6) in the rat. Ligation of spinal nerves induced symptoms mimicking tactile allodynia, as indicated by a marked decrease of the hindlimb withdrawal threshold to mechanical stimulation. The axon reflex induced by antidromic electrical stimulation of nociceptive fibres innervating the plantar skin ipsilateral to the ligation was attenuated according to determination of extravasation response and blood flow response. Lidocaine block or transection of the sciatic nerve of the neuropathic limb did not induce any change in basal blood flow of the plantar skin. The results indicate that ligation of spinal nerves induces an attenuation of the axon reflex. This attenuation reflects a decrease in the efferent function of primary afferent nociceptors innervating the hypersensitive skin of the hindpaw. The attenuation of antidromically-induced vascular responses was not caused by overriding sympathetic activity, as indicated by lack of blood flow effects by lidocaine blocks or a transection of the sciatic nerve.
Collapse
Affiliation(s)
- Timo Kauppila
- Department of Physiology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | | | | | | | | |
Collapse
|
43
|
Ma QP. The expression of bradykinin B(1) receptors on primary sensory neurones that give rise to small caliber sciatic nerve fibres in rats. Neuroscience 2002; 107:665-73. [PMID: 11720789 DOI: 10.1016/s0306-4522(01)00387-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The bradykinin B(1) receptor has been considered as an important mediator for inflammatory pain. In the present study, we have investigated the fibre types of sciatic nerve primary sensory neurones that express B(1) receptors by retrograde tracing in combination with immunohistochemical staining, or double-immunohistochemical staining. Approximately 12% of the A-fibre dorsal root ganglion neurones, retrogradely labelled from an intra-sciatic nerve injection of fluorescein isothiocyanate-conjugated cholera toxin B subunit, were B(1) receptor-immunoreactive. Over 70% of the small diameter dorsal root ganglion neurones, retrogradely labelled from an intra-sciatic nerve injection of tetramethylrhodamine isothiocyanate-conjugated wheat germ agglutinin, were B(1) receptor-immunoreactive. Over 50% of the (predominantly non-peptidergic) C-fibre dorsal root ganglion neurones, retrogradely labelled from an intra-sciatic nerve injection of fluorescein isothiocyanate-conjugated Bandeiraea simplicifolia isolectin B4, were B(1) receptor-immunoreactive. When calcitonin gene-related peptide, which is contained mainly in small caliber C- and A(delta)-fibre primary afferents, and B(1) receptors were stained with a double-immunofluorescent method, over 80% of the calcitonin gene-related peptide-positive dorsal root ganglion neurones were B(1) receptor-immunoreactive. From these results we suggest that B(1) receptors are predominantly expressed by small diameter primary afferent neurones that give rise to sciatic nerve fibres, which include both peptidergic and non-peptidergic C-fibres and A(delta)-fibres. Since peripheral nociceptive information is primarily transmitted by C- and A(delta)-fibres, B(1) receptors may be involved in the modulation of nociceptive transduction or transmission.
Collapse
Affiliation(s)
- Q P Ma
- Department of Pharmacology, Merck Sharp & Dohme Research Laboratories, Neuroscience Research Centre, Terlings Park, Harlow CM20 2QR, UK.
| |
Collapse
|
44
|
Seike M, Ikeda M, Morimoto A, Matsumoto M, Kodama H. Increased synthesis of calcitonin gene-related peptide stimulates keratinocyte proliferation in murine UVB-irradiated skin. J Dermatol Sci 2002; 28:135-43. [PMID: 11858952 DOI: 10.1016/s0923-1811(01)00155-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Repeated ultraviolet (UV) irradiations have been shown to induce keratinocyte proliferation with acanthosis, stimulate the cutaneous nerve proliferation, and increase the synthesis of calcitonin gene-related peptide (CGRP). In the current study, we examined the role of CGRP in the UVB-induced proliferation of murine keratinocytes. UVB irradiation increased the number of bromodeoxyuridine (BrdU)-labeled basal keratinocytes and caused acanthosis. In addition, CGRP expression was up-regulated in the peripheral nerves of the upper dermis and lower epidermis. Repeated intradermal injections of CGRP increased the number of BrdU-labeled basal cells and caused acanthosis. Intradermal injections of capsaicin prior to UVB-irradiation inhibited the UVB-induced CGRP expression, BrdU labeling in basal keratinocytes and epidermal thickening. Intradermal injections of anti-CGRP antibody inhibited the UVB-induced BrdU labeling in basal keratinocytes, but epidermal thickening was not significantly inhibited. These results indicate that CGRP is one of the stimulators to UVB-induced keratinocyte proliferation. On the other hand, expression of substance P, another neuropeptide in the peripheral nerve, was not up-regulated by UVB irradiation.
Collapse
Affiliation(s)
- Masahiro Seike
- Department of Dermatology, Kochi Medical School, Okohcho, Nankoku, 783-8505, Kochi, Japan.
| | | | | | | | | |
Collapse
|
45
|
Zhang L, Hoff AO, Wimalawansa SJ, Cote GJ, Gagel RF, Westlund KN. Arthritic calcitonin/alpha calcitonin gene-related peptide knockout mice have reduced nociceptive hypersensitivity. Pain 2001; 89:265-73. [PMID: 11166483 DOI: 10.1016/s0304-3959(00)00378-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peripheral inflammation induced with a knee joint injection of a mixture of kaolin/carrageenan (k/c) produces primary and secondary hyperalgesia. Inflammatory pain is thought to involve a variety of transmitters released from nerve terminals, including amino acids, substance P (SP) and calcitonin gene-related peptide (CGRP). In the present study, mice deficient in the calcitonin/alpha CGRP gene (CGRP(-/-)) displayed normal responses to noxious stimuli. However, the CGRP knockout mice failed to demonstrate development of secondary hyperalgesia after induction of knee joint inflammation in two tests that assess central sensitization, through testing at sites remote from the primary insult. Nociceptive behavioral responses were assessed using the hot-plate test and paw withdrawal latency (PWL) to radiant heat applied to the hindpaw. The CGRP(-/-) mice showed no signs of secondary hyperalgesia after development of knee joint inflammation, while the expected significant decrease in the PWL was observed in the CGRP(+/+) mice as control. The CGRP(-/-) mice also had a prolonged rather than a shortened response latency in the hot-plate test 4 h after knee joint injection of k/c. Immunohistological study showed that CGRP-like immunoreactivity (CGRP-LI) was absent in the spinal cord and dorsal root ganglia taken from the CGRP(-/-) mice. These results indicate that endogenous CGRP plays an important role in the plastic neurogenic changes occurring in response to peripheral inflammatory events including the development of nociceptive behaviors.
Collapse
Affiliation(s)
- L Zhang
- Department of Anatomy and Neurosciences, The University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, TX 77555-1069, USA
| | | | | | | | | | | |
Collapse
|
46
|
Ulfhake B, Bergman E, Edstrom E, Fundin BT, Johnson H, Kullberg S, Ming Y. Regulation of neurotrophin signaling in aging sensory and motoneurons: dissipation of target support? Mol Neurobiol 2000; 21:109-35. [PMID: 11379795 DOI: 10.1385/mn:21:3:109] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A hallmark of senescence is sensorimotor impairment, involving locomotion and postural control as well as fine-tuned movements. Sensory and motoneurons are not lost to any significant degree with advancing age, but do show characteristic changes in gene-expression pattern, morphology, and connectivity. This review covers recent experimental findings corroborating that alterations in trophic signaling may induce several of the phenotypic changes seen in primary sensory and motoneurons during aging. Furthermore, the data suggests that target failure, and/or breakdown of neuron-target interaction, is a critical event in the aging process of sensory and motoneurons.
Collapse
Affiliation(s)
- B Ulfhake
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
47
|
Hikiji A, Yamamoto H, Sunakawa M, Suda H. Increased blood flow and nerve firing in the cat canine tooth in response to stimulation of the second premolar pulp. Arch Oral Biol 2000; 45:53-61. [PMID: 10669092 DOI: 10.1016/s0003-9969(99)00114-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mustard oil or mechanical stimulation was applied to maxillary second premolar tooth pulps and pulpal blood flow and or intradental nerve activity in the ipsilateral canine tooth were recorded in the cat. Mustard oil application to the second premolar pulp significantly increased blood flow in the canine tooth pulp to 162.0+/-65.8% (n = 16) of the prestimulation flow compared to control data obtained with application of mineral oil (107.0+/-5.1%, n = 6) (Mann-Whitney U-test, p = 0.0009). Sectioning of the infraorbital nerve and its branches on the experimental side (n = 4) did not affect this increase in pulpal blood flow. The paraperiosteal injection of 2% lidocaine (1.0 ml) without vasoconstrictor significantly inhibited the increase in canine pulpal blood flow induced by mustard oil application to the second premolar pulp (109.8+/-6.8% of the prestimulation level, n = 7) (Mann-Whitney U-test, p = 0.0013). Sporadic firing or sometimes bursts of action potentials in the canine pulp nerves were recorded during and/or after the mustard oil application to the second premolar pulp in three of 16 cases. Four single pulp nerve units firing in synchrony with the mechanical stimulation of the second premolar pulp were recorded in two of eight canines, which substantiated the existence of branched afferents innervating both teeth. These findings suggest that stimulation of the second premolar pulp may induce axon reflex-related vasodilation and intradental nerve firing in the canine pulp via branched afferent fibres innervating both the second premolar and canine teeth.
Collapse
Affiliation(s)
- A Hikiji
- Department of Endodontics, Faculty of Dentistry, Tokyo Medical and Dental University, Japan.
| | | | | | | |
Collapse
|
48
|
Häbler HJ, Timmermann L, Stegmann JU, Jänig W. Involvement of neurokinins in antidromic vasodilatation in hairy and hairless skin of the rat hindlimb. Neuroscience 1999; 89:1259-68. [PMID: 10362313 DOI: 10.1016/s0306-4522(98)00322-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
By intravenous application of the specific neurokininl receptor antagonist SR 140333 and the specific calcitonin gene-related peptide receptor antagonist CGRP8-37 we tested to what extent neurokinins (substance P, neurokinin A) and calcitonin gene-related peptide are involved in mediating antidromic vasodilatation in skin of anaesthetized Wistar rats. The lumbar sympathetic chain was sectioned bilaterally between ganglia L2 and L3 to remove ongoing vasoconstrictor activity to the hindquarter. The left dorsal root L5 was stimulated electrically at 1 Hz with 20 pulses supramaximal for activating C-fibres to evoke antidromic vasodilatation which was measured with laser Doppler flowmetry on the glabrous plantar skin and the hairy skin of the lower hindlimb within the left L5 territory. Stimulation-induced vasodilatation was tested after applying SR 140333 (0.1 mg/kg) and CGRP8-37 (0.3 mg/kg) alone or in combination. SR 140333 delayed the onset of the vasodilatation, but did not change its amplitude. CGRP8-37 reduced the amplitude and duration of the vasodilatation, but did not affect the latency of its onset. In combination, SR 140333 potentiated the effect of CGRP8-37 on the amplitude of the vasodilatation in glabrous but not in hairy skin and CGRP8-37 potentiated the delayed onset produced by SR 140333 in both cutaneous tissues. Antidromic vasodilatation in glabrous skin was almost totally blocked by SR 140333 (0.1 mg/kg) in combination with CGRP8-37 (0.45 mg/kg), but a substantial dilatation remained in hairy skin. It is concluded that in rat glabrous skin the vasodilatation evoked by a low level of activity in small-diameter primary afferents is likely to result from the release and synergistic action of neurokinins (substance P and/or neurokinin A) and calcitonin gene-related peptide, while in hairy skin neurokinins are involved to a minor extent only.
Collapse
Affiliation(s)
- H J Häbler
- Physiologisches Institut, Christian-Albrechts-Universität, Kiel, Germany
| | | | | | | |
Collapse
|
49
|
Tonra JR. Classical and novel directions in neurotrophin transport and research: anterograde transport of brain-derived neurotrophic factor by sensory neurons. Microsc Res Tech 1999; 45:225-32. [PMID: 10383115 DOI: 10.1002/(sici)1097-0029(19990515/01)45:4/5<225::aid-jemt6>3.0.co;2-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
After the discovery of nerve growth factor, a classic model of neurotrophin action was developed. In this model, nerve endings compete for limited quantities of neurotrophic factors produced in neuronal target tissues. Neurotrophins are bound with high-affinity receptors expressed on the neuronal membrane and then endocytosed and retrogradely transported back to the cell body of responsive neurons. This classic model of target derived trophic support has been utilized to explain a wide range of trophic actions including effects on neuronal survival, terminal branching, and protein expression. However, a number of recent findings in the field of neurotrophin research cannot be explained using the classic model. In the peripheral nervous system (PNS), sensory neurons have been shown to contain mRNA for a member of the neurotrophin family, brain-derived neurotrophic factor (BDNF). Sensory neurons do not receive synaptic input so neurotrophin production by these cells does not fit into the classic target derived model. In contrast to target derived trophic support, BDNF produced by sensory neurons provides local autocrine and paracrine neurotrophic support in vitro. Furthermore, in vivo, sensory neurons transport BDNF in the anterograde direction away from the cell body, and opposite to the retrograde direction utilized in the classic model. Thus, out of necessity, a new direction for neurotrophin research has developed to study the production and anterograde transport of neurotrophins. The importance of this new mode of neurotrophin action in the PNS is indicated by results that implicate it in the response to pain, inflammation, and nerve injury.
Collapse
Affiliation(s)
- J R Tonra
- Millennium BioTherapeutics, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
50
|
Kress M, Guthmann C, Averbeck B, Reeh PW. Calcitonin gene-related peptide and prostaglandin E2 but not substance P release induced by antidromic nerve stimulation from rat skin in vitro. Neuroscience 1999; 89:303-10. [PMID: 10051237 DOI: 10.1016/s0306-4522(98)00280-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The liberation of calcitonin gene-related peptide from rat skin in vitro induced by antidromic electrical stimulation of unmyelinated units is demonstrated. Prostaglandin E2 was released concomitantly during C-fiber stimulation. A dose-dependent increase in prostaglandin E2 content of the eluate was also observed in response to stimulation with substance P (10(-7) to 10(-5) M) and calcitonin gene-related peptide (10(-6) and 10(-5) M). In contrast, prostaglandin E2 did not induce measurable release of neuropeptides. The amount of calcitonin gene-related peptide released during suprathreshold electrical stimulation increased with pulse frequency. Calcitonin gene-related peptide and prostaglandin release were completely inhibited in the presence of EMD 61753, a selective kappa-opioid receptor agonist. No significant release of substance P was observed. The data demonstrate a primary release of calcitonin gene-related peptide from unmyelinated but not myelinated primary afferents in the rat skin, which is accompanied by a secondary liberation of prostaglandin E2, connecting neurogenic inflammation to general mechanisms of inflammation.
Collapse
Affiliation(s)
- M Kress
- Institute of Physiology and Experimental Pathophysiology, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | |
Collapse
|