151
|
Abstract
Neurogenic inflammation and ensuing pain can be modulated by inhibiting the function of primary afferent neurons. The best studied mechanism to accomplish such inhibition is the opioid system. Under inflammatory conditions, the anterograde axonal transport of opioid receptors from dorsal root ganglia toward the peripheral sensory nerve endings is augmented. The increased number of opioid receptors (among other mechanisms) leads to improved analgesic effects of exogenously administered ligands (eg, morphine) and of endogenous leukocyte-derived opioid peptides (eg, beta-endorphin). A current concept proposes that during inflammatory processes endogenous opioid peptides can be secreted from immunocytes, occupy peripheral opioid receptors on sensory nerve endings, and produce analgesia by inhibiting the excitability of these nerves or the release of proinflammatory neuropeptides. This article focuses on the role of peripheral opioid receptors in pain control and on novel pharmaceutical concepts for the treatment of patients who suffer from rheumatoid arthritis and other inflammatory pain.
Collapse
Affiliation(s)
- Wiebke Puehler
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | |
Collapse
|
152
|
Brack A, Rittner HL, Machelska H, Leder K, Mousa SA, Schäfer M, Stein C. Control of inflammatory pain by chemokine-mediated recruitment of opioid-containing polymorphonuclear cells. Pain 2004; 112:229-238. [PMID: 15561377 DOI: 10.1016/j.pain.2004.08.029] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Revised: 08/12/2004] [Accepted: 08/23/2004] [Indexed: 01/22/2023]
Abstract
Opioid-containing leukocytes can counteract inflammatory hyperalgesia. Under stress or after local injection of corticotropin releasing factor (CRF), opioid peptides are released from leukocytes, bind to opioid receptors on peripheral sensory neurons and mediate antinociception. Since polymorphonuclear cells (PMN) are the predominant opioid-containing leukocyte subpopulation in early inflammation, we hypothesized that PMN and their recruitment by chemokines are important for peripheral opioid-mediated antinociception at this stage. Rats were intraplantarly injected with complete Freund's adjuvant (CFA). Using flow cytometry, immunohistochemistry, and ELISA, leukocyte subpopulations, chemokine receptor (CXCR2) expression on opioid-containing leukocytes and the CXCR2 ligands keratinocyte-derived chemokine (KC), macrophage inflammatory protein-2 (MIP-2) and cytokine-induced neutrophil chemoattractant-2 (CINC-2) were quantified. Paw pressure threshold (PPT) was determined before and after intraplantar and subcutaneous injection of CRF with or without naloxone. PMN depletion was achieved by intravenous injection of an antiserum. Chemokines were blocked by intraplantar injection of anti-MIP-2 and/or anti-KC antiserum. We found that at 2 h post CFA (i) intraplantar but not subcutaneous injection of CRF produced dose-dependent and naloxone-reversible antinociception (P<0.05, ANOVA). (ii) Opioid-containing leukocytes in the paw and CRF-induced antinociception were reduced after PMN depletion (P<0.05, t-test). (iii) Opioid-containing leukocytes mostly expressed CXCR2. MIP-2 and KC, but not CINC-2 were detectable in inflamed but not in noninflamed tissue (P<0.05, ANOVA). (iv) Combined but not single blockade of MIP-2 and KC reduced the number of opioid-containing leukocytes and peripheral opioid-mediated antinociception (P<0.05, t-test; P>0.05, ANOVA). In summary, in early inflammation peripheral opioid-mediated antinociception is critically dependent on PMN and their recruitment by CXCR2 chemokines.
Collapse
Affiliation(s)
- Alexander Brack
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
153
|
|
154
|
Sekido R, Ishimaru K, Sakita M. Corticotropin-releasing factor and interleukin-1beta are involved in the electroacupuncture-induced analgesic effect on inflammatory pain elicited by carrageenan. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2004; 32:269-79. [PMID: 15315264 DOI: 10.1142/s0192415x04001928] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Electroacupuncture (EA) is used to relieve various kinds of pain. However, the mechanistic basis of electroacupuncture analgesia (EAA) in inflammatory pain remains unclear. In the present study, we investigated whether endogenous peripheral corticotropin-releasing factor (CRF) or interleukin-1beta (IL-1) participated in EAA during hyperalgesia elicited by carrageenan-induced inflammation. Carrageenan was subcutaneously administered by intraplantar (i.pl.) injection of the left hind paw to induce inflammation. Nociceptive thresholds were measured using the paw pressure threshold (PPT) (Randall Sellito Test). Rats received 3 Hz EA in the left anterior tibial muscles for 1 hour after carrageenan injection. The selective CRF antagonist, alpha-helical CRF, or the recombinant IL-1 receptor antagonist, IL-1ra, was administered by i.pl. injection of the inflamed paw or by intravenous (i.v.) injection 1 hour before EA. PPT decreased significantly 3 hours after carrageenan injection. This decrease persisted at least 24 hours after carrageenan injection. EA resulted in significant increases of PPT, moreover, PPT elevations lasted 24 hours after carrageenan injection. By contrast, PPT elevations produced by EA were dose-dependently antagonized by local i.pl. injection of alpha-helical CRF or IL-1ra. This PPT elevation was not influenced by i.v. injection of alpha-helical CRF or IL-1ra. These findings suggest that peripheral CRF or IL-1 participate in EAA during hyperalgesia. The release of CRF or IL-1 elicited by EA may trigger the release of opioid peptides within inflamed tissue which may activate peripheral opioid receptors and inhibit the pain.
Collapse
Affiliation(s)
- Reina Sekido
- Department of Surgery, Graduate School of Acupuncture and Moxibustion, Meiji University of Oriental Medicine, Kyoto, Japan.
| | | | | |
Collapse
|
155
|
Binder W, Mousa SA, Sitte N, Kaiser M, Stein C, Schäfer M. Sympathetic activation triggers endogenous opioid release and analgesia within peripheral inflamed tissue. Eur J Neurosci 2004; 20:92-100. [PMID: 15245482 DOI: 10.1111/j.1460-9568.2004.03459.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stress induces analgesia by mechanisms within and outside the brain. Here we show that the sympathetic nervous system is an essential trigger of intrinsic opioid analgesia within peripheral injured tissue. Noradrenaline, injected directly into inflamed hind paws of male Wistar rats, produced dose-dependent antinociception, reversible by alpha(1)-, alpha(2)- and beta(2)-antagonists. alpha(1)-, alpha(2)- and beta(2)-adrenergic receptors were demonstrated on beta-endorphin-containing immune cells and noradrenaline induced adrenergic receptor-specific release of beta-endorphin from immune cell suspensions. This antinociceptive effect of noradrenaline was reversed by micro - and delta-opioid antagonists as well as by anti-beta-endorphin. Stress-induced peripheral analgesia was abolished by chemical sympathectomy and by adrenergic antagonists. These findings indicate that sympathetic neuron-derived noradrenaline stimulates adrenergic receptors on inflammatory cells to release beta-endorphin, which induces analgesia via activation of peripheral opioid receptors.
Collapse
Affiliation(s)
- Waltraud Binder
- Department of Anesthesiology and Critical Care Medicine, Klinikum Benjamin Franklin, Freie Universität, Hindenburgdamm 30, D-12200 Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
156
|
Tegeder I, Geisslinger G. Opioids as modulators of cell death and survival--unraveling mechanisms and revealing new indications. Pharmacol Rev 2004; 56:351-69. [PMID: 15317908 DOI: 10.1124/pr.56.3.2] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids are powerful analgesics but also drugs of abuse. Because opioid addicts are susceptible to certain infections, opioids have been suspected to suppress the immune response. This was supported by the finding that various immune-competent cells express opioid receptors and undergo apoptosis when treated with opioid alkaloids. Recent evidence suggests that opioids may also effect neuronal survival and proliferation or migrating properties of tumor cells. A multitude of signaling pathways has been suggested to be involved in these extra-analgesic effects of opioids. Growth-promoting effects were found to be mediated through Akt and Erk signaling cascades. Death-promoting effects have been ascribed to inhibition of nuclear factor-kappaB, increase of Fas expression, p53 stabilization, cytokine and chemokine release, and activation of nitric oxide synthase, p38, and c-Jun-N-terminal kinase. Some of the observed effects were inhibited with opioid receptor antagonists or pertussis toxin; others were unaffected. It is still unclear whether these properties are mediated through typical opioid receptor activation and inhibitory G-protein-signaling. The present review tries to unravel controversial findings and provides a hypothesis that may help to integrate diverse results.
Collapse
Affiliation(s)
- Irmgard Tegeder
- Pharmazentrum Frankfurt, Institut für Klinische Pharmakologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt, Germany.
| | | |
Collapse
|
157
|
Hutchinson MR, La Vincente SF, Somogyi AA. In vitro opioid induced proliferation of peripheral blood immune cells correlates with in vivo cold pressor pain tolerance in humans: a biological marker of pain tolerance. Pain 2004; 110:751-755. [PMID: 15288416 DOI: 10.1016/j.pain.2004.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Revised: 05/13/2004] [Accepted: 05/20/2004] [Indexed: 11/24/2022]
Abstract
There is substantial evidence for bidirectional communication between the immune system and the central nervous system, as the cells and signalling molecules of the immune system influence many central nervous system functions, for instance nociception. Opioids, such as morphine, produce analgesia and numerous other central and peripheral effects including sedation and euphoria, while their effects on the immune system are wide-ranging. There is considerable interindividual variability in basal nociception and response to opioids, however, the physiological and biological mechanisms underlying this are unclear. Therefore, we investigated the relationship between the immune system and basal nociceptive thresholds, using the proliferative response of isolated peripheral blood mononuclear cells and cold pressor pain tolerance. Here we show that the percent increase in proliferation of peripheral immune cells from 13 healthy subjects incubated with morphine ex vivo is highly correlated with the subjects' tolerance to noxious cold stimuli (Pearson r = 0.92, P < 0.0001). These pilot data provide evidence of a novel objective biological marker of pain tolerance in humans, which also links the immune and opioid systems with basal pain tolerance.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Department of Clinical and Experimental Pharmacology, Level 5, Medical School North, University of Adelaide, Frome Rd, Adelaide, SA 5005, Australia
| | | | | |
Collapse
|
158
|
Likar R, Mousa SA, Philippitsch G, Steinkellner H, Koppert W, Stein C, Schäfer M. Increased numbers of opioid expressing inflammatory cells do not affect intra-articular morphine analgesia. Br J Anaesth 2004; 93:375-80. [PMID: 15247115 DOI: 10.1093/bja/aeh222] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Both locally expressed beta-endorphin (END) and low doses of morphine relieve pain within inflamed knee joints. Here we examined whether enhanced inflammation and END expression within the synovial tissue of patients undergoing arthroscopic knee surgery might shift the analgesic dose-response curve of intra-articular (i.a.) morphine. METHODS Following IRB approval and informed consent, patients were randomly assigned to the following i.a. treatments at the end of surgery: group I (n=39), isotonic saline; group II (n=40), 1 mg morphine hydrochloride; group III (n=48), 2 mg morphine hydrochloride; group IV (n=39), 4 mg morphine hydrochloride. Postoperative pain intensity was assessed by the visual analogue scale (VAS), by the time to first analgesic request and by the supplemental piritramide consumption. Synovial specimens from each patient were stained for the presence of inflammatory cells and END and were discriminated into groups with low versus high numbers of these cells. Differences between groups were statistically analyzed by chi(2), anova and mancova where appropiate. RESULTS Patient characteristics and VAS scores did not differ between groups. Total postoperative piritramide consumption decreased and the time to first analgesic request increased significantly with increasing doses of i.a. morphine (P<0.05, anova and linear regression). These dose-response relationships were not different between patients with low versus high numbers of inflammatory and END-containing synovial cells (P>0.05, mancova). CONCLUSIONS The dose-response relationship of i.a. morphine analgesia is not shifted by enhanced inflammation and END expression within synovial tissue. Thus, the presence of END within inflamed synovial tissue does not seem to interfere with i.a. morphine analgesia.
Collapse
Affiliation(s)
- R Likar
- Abteilung für Anaesthesiologie und Intensivmedizin, LKH Klagenfurt, St. Veiter-Strasse 47, A-9020 Klagenfurt, Austria
| | | | | | | | | | | | | |
Collapse
|
159
|
Rosenberger P, Mühlbauer E, Weissmüller T, Rommelspacher H, Sinha P, Wernecke KD, Finckh U, Rettig J, Kox WJ, Spies CD. Decreased proopiomelanocortin mRNA in lymphocytes of chronic alcoholics after intravenous human corticotropin releasing factor injection. Alcohol Clin Exp Res 2004; 27:1693-700. [PMID: 14634483 DOI: 10.1097/01.alc.0000095636.44770.55] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Alcohol abuse may involve an altered neuroendocrine response that mediates lymphocyte-derived proopiomelanocortin (POMC) production and inflammation. We investigated POMC messenger RNA (mRNA) expression in human lymphocytes ex vivo and their relation to plasma ACTH and immunoreactive beta-endorphin (IR-beta-EP) after intravenous injection of human corticotropin releasing factor (hCRF) in chronic alcoholics (n = 12) and nonalcoholics (n = 12) before surgery. Lipopolysaccharide-stimulated interleukin (IL)-1 receptor antagonist (IL-1 Ra) as a marker for chronic inflammation was determined. METHODS Chronic alcohol abuse was diagnosed according to DSM-IV criteria and alcohol consumption >60 g/day. A reverse transcription-polymerase chain reaction method with total RNA and subsequent solid phase minisequencing was used to quantify lymphocytic POMC mRNA after intravenous hCRF injection. Plasma ACTH, cortisol, and lipopolysaccharide-stimulated IL-1 Ra of monocytes were measured by enzyme-linked immunosorbent assay, and plasma IR-beta-EP was measured by using radioimmunoassay. RESULTS Baseline values of POMC mRNA content in lymphocytes and IL-1 Ra of chronic alcoholics were significantly increased compared with nonalcoholics (p < 0.01). Thirty minutes after intravenous hCRF injection, a significant increase of lymphocytic POMC mRNA was measured (p < 0.05) in nonalcoholics, whereas in chronic alcoholics a significant decrease was observed (p < 0.05). CONCLUSIONS Chronic alcoholic patients had an altered neuroendocrine immune axis before intravenous hCRF administration and were not able to adjust to intravenous CRF injection by increasing lymphocytic POMC mRNA expression.
Collapse
Affiliation(s)
- Peter Rosenberger
- Department of Anesthesiology and Intensive Care, University Hospital Charité, Humboldt-University of Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Brack A, Rittner HL, Machelska H, Shaqura M, Mousa SA, Labuz D, Zöllner C, Schäfer M, Stein C. Endogenous peripheral antinociception in early inflammation is not limited by the number of opioid-containing leukocytes but by opioid receptor expression. Pain 2004; 108:67-75. [PMID: 15109509 DOI: 10.1016/j.pain.2003.12.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Revised: 11/21/2003] [Accepted: 12/01/2003] [Indexed: 11/27/2022]
Abstract
Endogenous inhibition of inflammatory pain is mediated by leukocytes that secrete opioid peptides upon exposure to stress (cold water swim stress, CWS) or after local injection of corticotropin releasing factor (CRF). Since in early inflammation few opioid-containing leukocytes are detected and since peripheral opioid-mediated antinociception is low we examined whether antinociception could be augmented by increased recruitment of opioid-containing polymorphonuclear cells (PMN). Rats were intraplantarly (i.pl.) injected with Freund's complete adjuvant (FCA) and with the PMN-recruiting chemokine macrophage inflammatory protein-2 (MIP-2, 1-10 microg; control: saline) for 2 h. Intraplantar leukocytes were quantified by flow cytometry. Paw pressure threshold (PPT) was determined before and after exposure to CWS, i.pl. injection of CRF and opioid peptides. Opioid receptors (OR) were measured by binding studies in dorsal root ganglia (DRG) and by immunohistochemistry in the paw. Our studies showed that (i) MIP-2 injection dose-dependently augmented recruitment of PMN and opioid-containing leukocytes (5-fold increase in cells/paw, P < 0.05), (ii) PPT was not different between groups at baseline and after CWS or CRF (maximum MPE: 20+/-2.3-29+/-7.2%, P < 0.05), (iii) injection of opioid peptides dose-dependently increased the PPT (P < 0.05, maximum MPE: and 18+/-2.6-21+/-3.6%), (iv) MOR (micro OR, MOP) binding sites in the ipsilateral DRG were unchanged (24+/-2-22+/-1.2 fmol/mg protein, P < 0.05, ANOVA) and (v) the number of MOR and DOR (delta OR, DOP) stained nerve fibers in peripheral tissue were unaltered (both P > 0.05, t-test). In summary, antinociception during early inflammation is apparently not limited by the number of opioid-containing leukocytes but by OR availability.
Collapse
Affiliation(s)
- Alexander Brack
- Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Klinik für Anaesthesiologie und operative Intensivmedizin, Hindenburgdamm 30, D-12200 Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Machelska H, Brack A, Mousa SA, Schopohl JK, Rittner HL, Schäfer M, Stein C. Selectins and integrins but not platelet-endothelial cell adhesion molecule-1 regulate opioid inhibition of inflammatory pain. Br J Pharmacol 2004; 142:772-80. [PMID: 15159283 PMCID: PMC1575048 DOI: 10.1038/sj.bjp.0705837] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Control of inflammatory pain can result from activation of opioid receptors on peripheral sensory nerves by opioid peptides secreted from leukocytes in response to stress (e.g. experimental swim stress or surgery). The extravasation of immunocytes to injured tissues involves rolling, adhesion and transmigration through the vessel wall, orchestrated by various adhesion molecules. 2. Here we evaluate the relative contribution of selectins, integrins alpha(4) and beta(2), and platelet-endothelial cell adhesion molecule-1 (PECAM-1) to the opioid-mediated inhibition of inflammatory pain. 3. We use flow cytometry, double immunofluorescence and nociceptive (paw pressure) testing in rats with unilateral hind paw inflammation induced by complete Freund's adjuvant. 4. In inflamed tissue, 43-58% of hematopoietic cells (CD45(+)) expressed opioid peptides. L-selectin and beta(2) were coexpressed by 7 and 98% of opioid-containing leukocytes, respectively. Alpha(4) integrin was expressed in low levels by the majority of leukocytes. Opioid-containing cells, vascular P- and E-selectin and PECAM-1 were simultaneously upregulated. 5. Swim stress produced potent opioid-mediated antinociception in inflamed tissue, unaffected by blockade of PECAM-1. However, blockade of L- and P-selectins by fucoidin, or of alpha(4) and beta(2) by monoclonal antibodies completely abolished peripheral stress-induced antinociception. This coincided with a 40% decrease in the migration of opioid-containing leukocytes to inflamed tissue. 6. These findings establish selectins and integrins alpha(4) and beta(2), but not PECAM-1, as important molecules involved in stress-induced opioid-mediated antinociception in inflammation. They point to a cautious use of anti-inflammatory treatments applying anti-selectin, anti-alpha(4) and anti-beta(2) strategies because they may impair intrinsic pain inhibition.
Collapse
Affiliation(s)
- Halina Machelska
- Klinik fur Anaesthesiologie und operative Intensivmedizin, Charite-Universitatsmedizin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
162
|
Hermanussen S, Do M, Cabot PJ. Reduction of ??-Endorphin-Containing Immune Cells in Inflamed Paw Tissue Corresponds with a Reduction in Immune-Derived Antinociception: Reversible by Donor Activated Lymphocytes. Anesth Analg 2004; 98:723-9, table of contents. [PMID: 14980927 DOI: 10.1213/01.ane.0000099369.23397.d7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
UNLABELLED The functional integrity of the immune system is essential for peripheral antinociception. Previous studies have demonstrated that immune cells elicit potent antinociception in inflamed tissues and that corticotropin-releasing factor-induced antinociception is significantly inhibited in animals that have undergone cyclosporin A (CsA)-induced immunosuppression. In this study, we examined the effect of a single bolus of CsA on inflammatory nociception. CsA-treated rats had substantially increased nociception compared with nonimmunosuppressed rats, consistent with a reduction in circulating and infiltrating lymphocytes. Furthermore, CsA-treated rats had inhibition of corticotropin-releasing factor-induced immune-derived antinociception, which was dose-dependently reversed by IV injection of concanavalin A-activated donor lymphocytes (1.0-7.0 x 10(6) cells/0.1 mL). In conclusion, our findings provided further evidence that opioid-containing immune cells are essential for peripheral analgesia. It is evident from these findings that control of inflammatory pain relies heavily on a functioning immune system. IMPLICATIONS The immune system not only contributes to inflammation, but also provides localized analgesia. A depleted immune system results in a reduction of immune-derived analgesia and a potentiation of inflammatory pain. Donor activated lymphocytes reverse these effects, highlighting the importance of a functional immune system in inflammatory pain.
Collapse
|
163
|
Mousa SA, Shakibaei M, Sitte N, Schäfer M, Stein C. Subcellular pathways of beta-endorphin synthesis, processing, and release from immunocytes in inflammatory pain. Endocrinology 2004; 145:1331-41. [PMID: 14630714 DOI: 10.1210/en.2003-1287] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The opioid peptide beta-endorphin (END) as well as mRNA for its precursor proopiomelanocortin (POMC) are found not only in the pituitary gland, but also within various types of immune cells infiltrating inflamed sc tissue. During stressful stimuli END is released and interacts with peripheral opioid receptors to inhibit pain. However, the subcellular pathways of POMC processing and END release have not yet been delineated in inflammatory cells. The aim of the present study was to examine the presence of POMC, carboxypeptidase E, the prohormone convertases 1 (PC1), and 2 (PC2), PC2-binding protein 7B2, and the release of END from inflammatory cells in rats. Using immunohistochemistry we detected END and POMC alone or colocalized with PC1, PC2, carboxypeptidase E, and 7B2 in macrophages/monocytes, granulocytes, and lymphocytes of the blood and within inflamed sc paw tissue. Immunoelectron microscopy revealed that END is localized within secretory granules packed in membranous structures in macrophages, monocytes, granulocytes, and lymphocytes. Finally, END is released by noradrenaline from immune cells in vitro. Taken together, our results indicate that immune cells express the entire machinery required for POMC processing into functionally active peptides such as END and are able to release these peptides from secretory granules.
Collapse
Affiliation(s)
- Shaaban A Mousa
- Klinik für Anaesthesiologie und Operative Intensivmedizin, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, D-12200 Berlin, Germany.
| | | | | | | | | |
Collapse
|
164
|
Mousa SA, Bopaiah PC, Stein C, Schäfer M. Involvement of corticotropin-releasing hormone receptor subtypes 1 and 2 in peripheral opioid-mediated inhibition of inflammatory pain. Pain 2003; 106:297-307. [PMID: 14659512 DOI: 10.1016/s0304-3959(03)00302-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In painful inflammation, exogenous as well as endogenous corticotropin-releasing hormone (CRH) can release opioid peptides (mainly beta-endorphin) from various types of immune cells and produce antinociception by activating opioid receptors on peripheral sensory nerve endings. CRH mediates its central effects through two high-affinity membrane receptors, the CRH receptor subtypes 1 and 2. It is unclear at present whether the peripheral antinociceptive effects of CRH are mediated through CRH receptor 1 (CRH R1) or CRH receptor 2 (CRH R2). Employing a double-immunocytochemical technique, this study investigated in Wistar rats with Freund's complete adjuvant-induced hind paw inflammation whether immune cells within blood and inflamed subcutaneous tissue express CRH R1 and/or CRH R2 together with the opioid peptide beta-endorphin (END). Additionally, we examined using selective CRH R1 and CRH R2 antagonists whether peripheral CRH-induced antinociception is mediated by the respective CRH receptor subtypes. We found a high degree of co-expression of END together with both CRH R1 and CRH R2 in macrophage/monocytes, granulocytes and lymphocytes within blood and inflamed subcutaneous tissue. Also we observed a high degree of co-localization of CRH R1 and CRH R2 receptors on circulating and resident immune cells. Both the selective CRH R1 antagonist CP-154,526 and the selective CRH R2 antagonist astressin 2B significantly attenuated peripheral antinociceptive effects of CRH indicating the involvement of both CRH receptor subtypes. Taken together, these findings suggest that in inflammatory pain CRH-induced peripheral antinociception is mediated via both CRH R1 and CRH R2 located on END containing immune cells within inflamed sites.
Collapse
Affiliation(s)
- Shaaban A Mousa
- Klinik für Anaesthesiologie und operative Intensivmedizin, Universitätsklinikum Benjamin Franklin, Freie Universität Berlin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | | | | | | |
Collapse
|
165
|
Szabo I, Wetzel MA, Zhang N, Steele AD, Kaminsky DE, Chen C, Liu-Chen LY, Bednar F, Henderson EE, Howard OMZ, Oppenheim JJ, Rogers TJ. Selective inactivation of CCR5 and decreased infectivity of R5 HIV-1 strains mediated by opioid-induced heterologous desensitization. J Leukoc Biol 2003; 74:1074-82. [PMID: 12972507 DOI: 10.1189/jlb.0203067] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The opiates are well-established immunomodulatory factors, and recent evidence suggests that mu- and delta-opioid receptor ligands alter chemokine-driven chemotactic responses through the process of heterologous desensitization. In the present report, we sought to examine the capacity of mu- and delta-opioids to modulate the function of chemokine receptors CCR5 and CXCR4, the two major human immunodeficiency virus (HIV) coreceptors. We found that the chemotactic responses to the CCR1/5 ligand CCL5/regulated on activation, normal T expressed and secreted, but not the CXCR4 ligand stromal cell-derived factor-1alpha/CXCL12 were inhibited following opioid pretreatment. Studies were performed with primary monocytes and Chinese hamster ovary cells transfected with CCR5 and the micro-opioid receptor to determine whether cross-desensitization of CCR5 was a result of receptor internalization. Using radiolabeled-binding analysis, flow cytometry, and confocal microscopy, we found that the heterologous desensitization of CCR5 was not associated with a significant degree of receptor internalization. Despite this, we found that the cross-desensitization of CCR5 by opioids was associated with a decrease in susceptibility to R5 but not X4 strains of HIV-1. Our findings are consistent with the notion that impairment of the normal signaling activity of CCR5 inhibits HIV-1 coreceptor function. These results have significant implications for our understanding of the effect of opioids on the regulation of leukocyte trafficking in inflammatory disease states and the process of coreceptor-dependent HIV-1 infection. The interference with HIV-1 uptake by heterologous desensitization of CCR5 suggests that HIV-1 interaction with this receptor is not passive but involves a signal transduction process.
Collapse
MESH Headings
- Acquired Immunodeficiency Syndrome/prevention & control
- Animals
- CHO Cells
- Calcium/metabolism
- Chemokine CXCL12
- Chemokines, CXC/metabolism
- Chemotaxis
- Cricetinae
- Desensitization, Immunologic
- Disease Susceptibility
- HIV Core Protein p24/genetics
- HIV Long Terminal Repeat
- HIV-1/physiology
- Humans
- Jurkat Cells
- Monocytes/metabolism
- Receptors, CCR5/genetics
- Receptors, CCR5/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, mu/physiology
- Signal Transduction
- Transfection
- Virus Replication/drug effects
- Virus Replication/physiology
Collapse
Affiliation(s)
- Imre Szabo
- Department of Microbiology and Immunology, Temple University School of Medicine, 3400 N. Broad Street, Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Kraus J, Börner C, Giannini E, Höllt V. The role of nuclear factor kappaB in tumor necrosis factor-regulated transcription of the human mu-opioid receptor gene. Mol Pharmacol 2003; 64:876-84. [PMID: 14500744 DOI: 10.1124/mol.64.4.876] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids and their receptors are key players in a cross-talk between the nervous and immune systems. For example, the endogenous opioid system is activated during inflammation as a physiological feedback mechanism to attenuate inflammatory pain. Herein, we report that in primary human T lymphocytes, Raji B cells, U937 monocytes, primary human polymorphonuclear leukocytes, and mature dendritic cells, the proinflammatory cytokine tumor necrosis factor induced mu-opioid receptor gene transcription. Transcriptional induction of the gene in immune cells was mediated via tumor necrosis factor receptor type 2. Using selective in vivo disruption of possibly involved transcription factors with decoy oligonucleotides, nuclear factor-kappaB was identified as the factor responsible for induction of the gene in immune cells, whereas activator protein-1 was found to be uninvolved. Nuclear factor-kappaB also mediates up-regulation of mu-opioid receptors in neuronal cells stimulated with tumor necrosis factor. Among six putative nuclear factor-kappaB binding sites on the mu-opioid receptor gene promoter, three cis-active elements at nt -2174, -557, and -207 were identified using transfection experiments of reporter gene constructs, electrophoretic mobility shift assays, and in vivo binding studies with decoy oligonucleotides. An allelic variation within the -557 element significantly reduced its trans-activating potency, which may affect regulation of the mu-opioid receptor gene in persons carrying this mutation. This study suggests a regulatory function of tumor necrosis factor in opioid-mediated processes in neuronal and immune cells, with possible impact on the complex of inflammation-induced analgesia.
Collapse
MESH Headings
- Antigens, CD/physiology
- Binding Sites
- Humans
- NF-kappa B/physiology
- Promoter Regions, Genetic/physiology
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type II
- Transcription Factor AP-1/metabolism
- Transcription, Genetic
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Jürgen Kraus
- Department of Pharmacology and Toxicology, University of Magdeburg, 44 Leipziger Strasse, 39120 Magdeburg, Germany.
| | | | | | | |
Collapse
|
167
|
Csaba G, Kovács P, Pállinger E. Endorphin content of white blood cells and peritoneal cells in neonatally benzpyrene treated adult rats. ACTA ACUST UNITED AC 2003; 90:207-15. [PMID: 14594191 DOI: 10.1556/aphysiol.90.2003.3.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
White blood cells of rats (lymphocytes, monocytes, macrophages, granulocytes and mast cells) contain beta-endorphin. Two months after a single neonatal benzpyrene treatment (imprinting) there is an elevated level of immunoreactive endorphin in the blood and peritoneal cells of female animals and blood cells of males. The endorphin content decreased in the peritoneal cells of males. In the blood, the granulocytes of female, and the lymphocytes of male rats contained the highest amount of endorphin. In the peritoneal fluid also the granulocytes of females contained the highest amount of endorphin, in contrast to males, where the endorphin content of cells decreased and the lowest level of it was present in the lymphocytes. The experiments justify that benzpyrene treatment can durably influence endorphin levels of white blood cells and gives new data to the already known lifelong health destroying effects of perinatal benzpyrene exposition (alterations of hormone receptor binding capacity and sexual behavior).
Collapse
Affiliation(s)
- G Csaba
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary.
| | | | | |
Collapse
|
168
|
Machelska H, Schopohl JK, Mousa SA, Labuz D, Schäfer M, Stein C. Different mechanisms of intrinsic pain inhibition in early and late inflammation. J Neuroimmunol 2003; 141:30-9. [PMID: 12965251 DOI: 10.1016/s0165-5728(03)00213-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neuroimmune interactions control pain through activation of opioid receptors on sensory nerves by immune-derived opioid peptides. Here we evaluate mechanisms of intrinsic pain inhibition at different stages of Freund's adjuvant-induced inflammation of the rat paw. We use immunohistochemistry and paw pressure testing. Our data show that in early (6 h) inflammation leukocyte-derived beta-endorphin, met-enkephalin and dynorphin A activate peripheral mu-, delta- and kappa-receptors to inhibit nociception. In addition, central opioid mechanisms seem to contribute significantly to this effect. At later stages (4 days), antinociception is exclusively produced by leukocyte-derived beta-endorphin acting at peripheral mu and delta receptors. Corticotropin-releasing hormone (CRH) is an endogenous trigger of these effects at both stages. These findings indicate that peripheral opioid mechanisms of pain inhibition gain functional relevance with the chronicity of inflammation.
Collapse
Affiliation(s)
- Halina Machelska
- Klinikum Benjamin Franklin, Freie Universität Berlin, D-12200 Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
169
|
Abstract
The treatment of severe pain with opioids has thus far been limited by their unwanted central side effects. Recent research promises new approaches, including opioid analgesics acting outside the central nervous system, targeting of opioid peptide-containing immune cells to peripheral damaged tissue, and gene transfer to enhance opioid production at sites of injury.
Collapse
Affiliation(s)
- Christoph Stein
- Klinik für Anaesthesiologie und Operative Intensivmedizin, Freie Universität Berlin, Klinikum Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | | | |
Collapse
|
170
|
Philippe D, Dubuquoy L, Groux H, Brun V, Chuoï-Mariot MTV, Gaveriaux-Ruff C, Colombel JF, Kieffer BL, Desreumaux P. Anti-inflammatory properties of the mu opioid receptor support its use in the treatment of colon inflammation. J Clin Invest 2003. [PMID: 12727924 DOI: 10.1172/jci200316750] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The physiologic role of the mu opioid receptor (MOR) in gut nociception, motility, and secretion is well established. To evaluate whether MOR may also be involved in controlling gut inflammation, we first showed that subcutaneous administration of selective peripheral MOR agonists, named DALDA and DAMGO, significantly reduces inflammation in two experimental models of colitis induced by administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) or peripheral expansion of CD4(+) T cells in mice. This therapeutic effect was almost completely abolished by concomitant administration of the opioid antagonist naloxone. Evidence of a genetic role for MOR in the control of gut inflammation was provided by showing that MOR-deficient mice were highly susceptible to colon inflammation, with a 50% mortality rate occurring 3 days after TNBS administration. The mechanistic basis of these observations suggests that the anti-inflammatory effects of MOR in the colon are mediated through the regulation of cytokine production and T cell proliferation, two important immunologic events required for the development of colon inflammation in mice and patients with inflammatory bowel disease (IBD). These data provide evidence that MOR plays a role in the control of gut inflammation and suggest that MOR agonists might be new therapeutic molecules in IBD.
Collapse
Affiliation(s)
- David Philippe
- Equipe Mixte INSERM 0114 sur la Physiopathologie des Maladies Inflammatoires Intestinales, Centre Hospitalier Universitaire, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Machelska H, Stein C. Peripheral Opioid Analgesia Neuroimmune Interactions and Therapeutic Implications. Pain 2003. [DOI: 10.1201/9780203911259.ch33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
172
|
DeHaven-Hudkins D. Profile of ADL 2-1294, an Opioid Antihyperalgesic Agent with Selectivity for Peripheral Mu-Opiate Receptors. Pain 2003. [DOI: 10.1201/9780203911259.ch34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
173
|
Philippe D, Dubuquoy L, Groux H, Brun V, Chuoï-Mariot MTV, Gaveriaux-Ruff C, Colombel JF, Kieffer BL, Desreumaux P. Anti-inflammatory properties of the mu opioid receptor support its use in the treatment of colon inflammation. J Clin Invest 2003; 111:1329-38. [PMID: 12727924 PMCID: PMC154442 DOI: 10.1172/jci16750] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The physiologic role of the mu opioid receptor (MOR) in gut nociception, motility, and secretion is well established. To evaluate whether MOR may also be involved in controlling gut inflammation, we first showed that subcutaneous administration of selective peripheral MOR agonists, named DALDA and DAMGO, significantly reduces inflammation in two experimental models of colitis induced by administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) or peripheral expansion of CD4(+) T cells in mice. This therapeutic effect was almost completely abolished by concomitant administration of the opioid antagonist naloxone. Evidence of a genetic role for MOR in the control of gut inflammation was provided by showing that MOR-deficient mice were highly susceptible to colon inflammation, with a 50% mortality rate occurring 3 days after TNBS administration. The mechanistic basis of these observations suggests that the anti-inflammatory effects of MOR in the colon are mediated through the regulation of cytokine production and T cell proliferation, two important immunologic events required for the development of colon inflammation in mice and patients with inflammatory bowel disease (IBD). These data provide evidence that MOR plays a role in the control of gut inflammation and suggest that MOR agonists might be new therapeutic molecules in IBD.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- CD4-Positive T-Lymphocytes/metabolism
- Colitis/chemically induced
- Colitis/drug therapy
- Colitis/mortality
- Colitis/pathology
- Colon/drug effects
- Colon/immunology
- Colon/pathology
- Cytokines/metabolism
- Disease Models, Animal
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Oligopeptides/pharmacology
- Oligopeptides/therapeutic use
- Peroxidase/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Trinitrobenzenesulfonic Acid/toxicity
Collapse
Affiliation(s)
- David Philippe
- Equipe Mixte INSERM 0114 sur la Physiopathologie des Maladies Inflammatoires Intestinales, Centre Hospitalier Universitaire, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Jessop DS. Neuropeptides: modulators of immune responses in health and disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 52:67-91. [PMID: 12498101 DOI: 10.1016/s0074-7742(02)52006-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Affiliation(s)
- David S Jessop
- University Research Center for Neuroendocrinology, University of Bristol, Bristol BS2 8HW, United Kingdom
| |
Collapse
|
175
|
Wacnik PW, Kehl LJ, Trempe TM, Ramnaraine ML, Beitz AJ, Wilcox GL. Tumor implantation in mouse humerus evokes movement-related hyperalgesia exceeding that evoked by intramuscular carrageenan. Pain 2003; 101:175-86. [PMID: 12507712 DOI: 10.1016/s0304-3959(02)00312-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In this paper we compare two innovative models of movement-related pain: tumor-induced nociception following implantation of fibrosarcoma cells into bone and muscle inflammation-induced nociception following injection of the irritant carrageenan into muscle. Importantly, using the grip force test, an assay of movement-related hyperalgesia, both non-malignant and malignant pain are examined in parallel. Movement-related hyperalgesia, known clinically as a specific type of 'breakthrough pain', is a common feature of bone cancer and is thought to be a predictor of poor response to conventional analgesic pharmacotherapy (Bruera et al., 1995, J. Pain Symptom. Manage. 10 (1995) 348; Mercadaute et al., 1992, Pain 50 (1992) 151; Pain 81 (1999) 129). Implantation of NCTC 2472 sarcoma cells in both humeri or injection of carrageenan (4%) in both triceps of C3H/He mice produced apparent forelimb hyperalgesia that was not associated with mechanical hyperalgesia in the forepaw, whereas carrageenan at 6 and 8% did evoke significant cutaneous hyperalgesia of the forepaw as well. Control groups receiving implants of vehicle or no treatment at all did not manifest this forelimb hyperalgesia. B6C3/F1 mice implanted with non-lysis-inducing G3.26 melanoma cells or vehicle did not manifest significant hyperalgesia when compared to B6C3/F1 mice receiving fibrosarcoma cells, indicating a dependence on bone involvement for induction of hyperalgesia in this model. Histological examination at days 3, 7, and 10 post-implantation showed a clear correlation of tumor growth-induced bone destruction with behavioral hyperalgesia. Morphine was more potent in decreasing the maximal hyperalgesia induced by carrageenan than that induced by tumor implantation. Acutely administered morphine (3-100mg/kg, i.p.) attenuated peak hyperalgesia of carrageenan-injected mice (ED(50) 6.9 mg/kg) and tumor-bearing mice (ED(50) 23.9 mg/kg) in a dose-related manner with a difference in potency of 3.5. Tumor-implanted mice with a level of hyperalgesia comparable to that induced by carrageenan required almost three times more morphine (ED(50) 18.5mg/kg) for equivalent attenuation of forelimb hyperalgesia. These animal models of movement-related hyperalgesia may aid in discerning the peripheral and central mechanisms underlying pain that accompanies bone metastases and distinguishing it from the pain associated with muscular inflammation. Importantly, they may also aid in predicting differences in analgesic efficacy in different types of musculoskeletal pain.
Collapse
Affiliation(s)
- Paul W Wacnik
- Department of Pharmacology, Medical School, University of Minnesota, 6-120 Jackson, 321 Church St SE, Minneapolis, MN 55455-0217, USA
| | | | | | | | | | | |
Collapse
|
176
|
Gavériaux-Ruff C, Simonin F, Filliol D, Kieffer BL. Enhanced humoral response in kappa-opioid receptor knockout mice. J Neuroimmunol 2003; 134:72-81. [PMID: 12507774 DOI: 10.1016/s0165-5728(02)00419-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opiates are major analgesics and addictive drugs described also as immunomodulators. Here, we investigated the contribution of kappa-opioid receptor (KOR) activity in immunity in vivo by studying immune responses in KOR knockout mice. These animals displayed a modest reduction in thymus cellularity and CD4(+) cell ratio, parallel to a slight increase in immature CD4(+)CD8(+) lymphocytes. In spleen, KOR null animals showed augmented cell number with no change in cell distribution. T and B lymphocyte proliferative capabilities in vitro, Natural Killer activity and steady-state Ig levels were unchanged in KOR-/- mice. We immunized the mice with the antigen keyhole limpet hemocyanin (KLH). Compared to wild-type (WT) mice, KOR-/- animals produced significant higher levels of antigen-specific total Ig, IgM, IgG1 and IgG2a antibodies. This enhancement of humoral activity was not observed in mu-opioid receptor and delta-opioid receptor knockout animals. These results show that endogenous activation of kappa-opioid receptors may exert a tonic inhibition of antibody (Ab) response.
Collapse
Affiliation(s)
- Claire Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, BP10142, 67404 Illkirch Cedex, C.U. de Strasbourg, France.
| | | | | | | |
Collapse
|
177
|
Dell'Antonio G, Quattrini A, Cin ED, Fulgenzi A, Ferrero ME. Relief of inflammatory pain in rats by local use of the selective P2X7 ATP receptor inhibitor, oxidized ATP. ARTHRITIS AND RHEUMATISM 2002; 46:3378-85. [PMID: 12483745 DOI: 10.1002/art.10678] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Oxidized ATP (oATP) is a selective inhibitor of the P2Z/P2X7 ATP receptor for extracellular ATP, which contributes to the antinociceptive effect. This study sought to determine the mechanism by which local administration of oATP is able to relieve inflammatory pain in arthritic rat paws. METHODS Arthritis was induced in Wistar rats by injections of Freund's complete adjuvant into one hind paw. Nociceptive thresholds were measured before and after local injection of oATP into the inflamed paws. The influence on pain transmission due to the presence of recruited inflammatory cells at the site of inflammation was determined by inhibiting the initial phase of their migration (by intravenous treatment with fucoidin, which blocks the adhesion molecules of the selectin family). ATP intraplantar content was determined in the different experimental conditions. Histologic features of the hind paws were evaluated by using the anti-P2X7 receptor polyclonal antibody. RESULTS Intraplantar administration of oATP into inflamed paws significantly relieved inflammatory pain. The antinociceptive effect of oATP was independent of the immune-cell recruitment. ATP levels in inflamed tissues were significantly reduced by oATP treatment. A variable presence of P2X7 receptors on cutaneous sensory nerves with respect to the different treatments was observed. Following oATP treatment, there was a reduction in P2X7 expression in the endings of peripheral nerves, as well as in endothelial cells. CONCLUSION Oxidized ATP inhibits inflammatory pain in arthritic rats by inhibition of the P2X7 receptor for ATP, which is localized on nerve terminals.
Collapse
|
178
|
Macé G, Jaume M, Blanpied C, Stephan L, Coudert JD, Druet P, Dietrich G. Anti-mu-opioid-receptor IgG antibodies are commonly present in serum from healthy blood donors: evidence for a role in apoptotic immune cell death. Blood 2002; 100:3261-8. [PMID: 12384426 DOI: 10.1182/blood-2002-01-0055] [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: 11/20/2022] Open
Abstract
We previously observed the presence of anti-human mu-opioid-receptor (anti-hMOR) autoantibodies in IgG pools prepared from several thousand healthy blood donors. These autoantibodies behaved agonistically because of their ability to bind to the first and third extracellular loops of the receptor. In this study, we found that each healthy donor's serum contained anti-hMOR IgG autoantibodies with a specific activity against both the first and the third extracellular loops of the receptor. Because of the inability of IgG to cross the blood-brain barrier, we investigated the effects of the expression of anti-hMOR autoantibodies on immune cells. In analogy to studies of the effects of morphine, we investigated the ability of antibodies to sensitize splenocytes to Fas (CD95)-mediated apoptosis. We took advantage of the high sequence homology between murine MOR and hMOR extracellular loops to estimate the effect on murine splenocytes of anti-hMOR antibodies raised by immunizing mice. Splenocytes from mice injected with Chinese hamster ovary (CHO) cells expressing MOR were sensitized to Fas-mediated apoptosis, whereas those from mice injected with CHO cells or phosphate-buffered saline were not. Similar sensitization to Fas-mediated apoptosis was observed in splenocytes from mice undergoing passive transfer either with IgG from mice previously immunized against CHO cells expressing MOR or with IgG directed against the first and third extracellular loops of the receptor. Together, our data show that anti-MOR autoantibodies are commonly expressed in healthy humans and could participate in the control of lymphocyte homeostasis by promoting Fas-mediated apoptosis.
Collapse
Affiliation(s)
- Gaëtane Macé
- Institut National de la Santé et de la Recherche Médicale (INSERM) U563, Department of Oncogenesis and Signaling in Haematopoietic Cells, Institut Claude de Préval, IFR 30, Hôpital Purpan, Faculté de Médecine Purpan, 31059 Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
179
|
Affiliation(s)
- Halina Machelska
- Klinik für Anaesthesiologie und operative Intensivmedizin, Klinikum Benjamin Franklin, Freie Universität Berlin, Germany.
| | | |
Collapse
|
180
|
|
181
|
Refojo D, Kovalovsky D, Young JI, Rubinstein M, Holsboer F, Reul JMHM, Low MJ, Arzt E. Increased splenocyte proliferative response and cytokine production in beta-endorphin-deficient mice. J Neuroimmunol 2002; 131:126-34. [PMID: 12458044 DOI: 10.1016/s0165-5728(02)00268-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We used beta-endorphin-deficient mice as a novel approach to confirm the physiological role that opioid peptides play in the development or regulation of the immune system. We found that mice lacking beta-endorphin possessed an enhanced immune response, measured in terms of splenocyte proliferation and interleukin (IL)-2 mRNA levels, in vitro production of the splenic macrophage inflammatory cytokines IL-6 and Tumor Necrosis Factor (TNF)-alpha and plasma IL-6 following lipopolysaccharide (LPS) administration. beta-Endorphin-deficient mice had attenuated increases of plasma ACTH and corticosterone levels in response to LPS. These results are consistent with a postulated inhibitory role of endogenous beta-endorphin on the immune system at multiple levels.
Collapse
Affiliation(s)
- Damian Refojo
- Laboratorio de Fisiología y Biología Molecular, Departamento de Biología, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Ciudad Universitaria, Pabellon II, C1428EHA, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
182
|
Local injection of a selective endothelin-B receptor agonist inhibits endothelin-1-induced pain-like behavior and excitation of nociceptors in a naloxone-sensitive manner. J Neurosci 2002. [PMID: 12196602 DOI: 10.1523/jneurosci.22-17-07788.2002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We showed previously that subcutaneous injection of the injury-associated peptide mediator endothelin-1 (ET-1) into the rat plantar hindpaw produces pain behavior and selective excitation of nociceptors, both through activation of ET(A) receptors likely on nociceptive terminals. The potential role of ET(B) receptor activation in these actions of ET-1-has not been examined. Therefore, in these experiments, we studied the effect of blocking or activating ET(B) receptors on ET-1-induced hindpaw flinching and excitation of nociceptors in rats. An ET(B) receptor-selective antagonist, BQ-788 (3 mm), coinjected with ET-1 (200 microm) reduced the time-to-peak of flinching and significantly enhanced the average maximal flinch frequency (MFF). In contrast, coinjection of an ET(B) receptor selective agonist, IRL-1620 (100 or 200 microm), with ET-1 reduced the average MFF and the average total number of flinches. Interestingly, this unexpected inhibitory effect of IRL-1620 was prevented by the nonselective opioid receptor antagonist naloxone (2.75 mm). To confirm these inhibitory actions, we studied the effects of IRL-1620 on ET-1-induced spike responses in single, physiologically characterized nociceptive C-fibers. IRL-1620 suppressed spike responses to ET-1 in all (n = 12) C-units, with mean and maximum response frequencies of 0.08 +/- 0.02 and 1.5 +/- 0.4 impulses/sec versus 0.32 +/- 0.07 and 4.17 +/- 0.17 impulses/sec for ET-1 alone. In additional support of the behavioral results, coinjection of naloxone (2.75 mm) completely prevented this inhibitory action of IRL-1620. These results establish that ET(B) receptor activation inhibits ET-1-induced pain behavior and nociception in a naloxone-sensitive manner and point to a previously unrecognized dual modulation of acute nociceptive signaling by ET(A) and ET(B) receptors in cutaneous tissues.
Collapse
|
183
|
Ippolito DL, Temkin PA, Rogalski SL, Chavkin C. N-terminal tyrosine residues within the potassium channel Kir3 modulate GTPase activity of Galphai. J Biol Chem 2002; 277:32692-6. [PMID: 12082117 PMCID: PMC1414899 DOI: 10.1074/jbc.m204407200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
trkB activation results in tyrosine phosphorylation of N-terminal Kir3 residues, decreasing channel activation. To determine the mechanism of this effect, we reconstituted Kir3, trkB, and the mu opioid receptor in Xenopus oocytes. Activation of trkB by BDNF (brain-derived neurotrophic factor) accelerated Kir3 deactivation following termination of mu opioid receptor signaling. Similarly, overexpression of RGS4, a GTPase-activating protein (GAP), accelerated Kir3 deactivation. Blocking GTPase activity with GTPgammaS also prevented Kir3 deactivation, and the GTPgammaS effect was not reversed by BDNF treatment. These results suggest that BDNF treatment did not reduce Kir3 affinity for Gbetagamma but rather acted to accelerate GTPase activity, like RGS4. Tyrosine phosphatase inhibition by peroxyvanadate pretreatment reversibly mimicked the BDNF/trkB effect, indicating that tyrosine phosphorylation of Kir3 may have caused the GTPase acceleration. Tyrosine to phenylalanine substitution in the N-terminal domain of Kir3.4 blocked the BDNF effect, supporting the hypothesis that phosphorylation of these tyrosines was responsible. Like other GAPs, Kir3.4 contains a tyrosine-arginine-glutamine motif that is thought to function by interacting with G protein catalytic domains to facilitate GTP hydrolysis. These data suggest that the N-terminal tyrosine hydroxyls in Kir3 normally mask the GAP activity and that modification by phosphorylation or phenylalanine substitution reveals the GAP domain. Thus, BDNF activation of trkB could inhibit Kir3 by facilitating channel deactivation.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- Arginine/chemistry
- Brain-Derived Neurotrophic Factor/pharmacology
- Catalytic Domain
- Enzyme Activation
- G Protein-Coupled Inwardly-Rectifying Potassium Channels
- GTP Phosphohydrolases/metabolism
- GTP-Binding Protein alpha Subunit, Gi2
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- GTP-Binding Protein alpha Subunits, Gi-Go/physiology
- Glutamine/chemistry
- Hydrolysis
- Kinetics
- Models, Chemical
- Patch-Clamp Techniques
- Potassium Channels/chemistry
- Potassium Channels/metabolism
- Potassium Channels, Inwardly Rectifying
- Protein Binding
- Protein Structure, Tertiary
- Protein Tyrosine Phosphatases/metabolism
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/physiology
- RGS Proteins/metabolism
- RNA, Complementary/metabolism
- Receptor, trkB/metabolism
- Receptors, Opioid, mu/metabolism
- Tyrosine/chemistry
- Tyrosine/metabolism
- Vanadates/pharmacology
- Xenopus
Collapse
Affiliation(s)
| | | | | | - Charles Chavkin
- ‡ To whom correspondence should be addressed: Dept. of Pharmacology, University of Washington, Box 357280, Seattle, WA 98195-7280. Tel.: 206-543-4266; Fax: 206-685-3822; E-mail:
| |
Collapse
|
184
|
Khodorova A, Fareed MU, Gokin A, Strichartz GR, Davar G. Local injection of a selective endothelin-B receptor agonist inhibits endothelin-1-induced pain-like behavior and excitation of nociceptors in a naloxone-sensitive manner. J Neurosci 2002; 22:7788-96. [PMID: 12196602 PMCID: PMC6757974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
We showed previously that subcutaneous injection of the injury-associated peptide mediator endothelin-1 (ET-1) into the rat plantar hindpaw produces pain behavior and selective excitation of nociceptors, both through activation of ET(A) receptors likely on nociceptive terminals. The potential role of ET(B) receptor activation in these actions of ET-1-has not been examined. Therefore, in these experiments, we studied the effect of blocking or activating ET(B) receptors on ET-1-induced hindpaw flinching and excitation of nociceptors in rats. An ET(B) receptor-selective antagonist, BQ-788 (3 mm), coinjected with ET-1 (200 microm) reduced the time-to-peak of flinching and significantly enhanced the average maximal flinch frequency (MFF). In contrast, coinjection of an ET(B) receptor selective agonist, IRL-1620 (100 or 200 microm), with ET-1 reduced the average MFF and the average total number of flinches. Interestingly, this unexpected inhibitory effect of IRL-1620 was prevented by the nonselective opioid receptor antagonist naloxone (2.75 mm). To confirm these inhibitory actions, we studied the effects of IRL-1620 on ET-1-induced spike responses in single, physiologically characterized nociceptive C-fibers. IRL-1620 suppressed spike responses to ET-1 in all (n = 12) C-units, with mean and maximum response frequencies of 0.08 +/- 0.02 and 1.5 +/- 0.4 impulses/sec versus 0.32 +/- 0.07 and 4.17 +/- 0.17 impulses/sec for ET-1 alone. In additional support of the behavioral results, coinjection of naloxone (2.75 mm) completely prevented this inhibitory action of IRL-1620. These results establish that ET(B) receptor activation inhibits ET-1-induced pain behavior and nociception in a naloxone-sensitive manner and point to a previously unrecognized dual modulation of acute nociceptive signaling by ET(A) and ET(B) receptors in cutaneous tissues.
Collapse
Affiliation(s)
- Alla Khodorova
- Molecular Neurobiology of Pain, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
185
|
Abstract
The distribution and concentration of leu-enkephalin in periosteum, cortical bone, bone marrow and synovial membrane of normal rats were analysed. Periosteum, cortical bone and bone marrow of the rat femurs were collected as well as the ankles. The distribution of leu-enkephalin was analysed by immunoelectron microscopy and the concentration of leu-enkephalin was measured with radioimmunoassay. Immunoelectron microscopy showed that leu-enkephalin is abundant in monocytes of bone marrow, nerve fibers and endothelial cells in the periosteum and also in macrophage-like-cells of the synovial membrane. The concentration of leu-enkephalin measured by RIA showed highest concentration in bone marrow followed by periosteum and cortical bone. The study supports that leu-enkephalin is present and can be quantified in different compartments of bone and joint tissues suggesting that leu-enkephalin may be involved in the physiological regulation of nociception and immunoregulation.
Collapse
Affiliation(s)
- Wu Qinyang
- Department of Orthopedic Surgery, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|
186
|
Abstract
Pain can be effectively controlled by endogenous mechanisms based on neuroimmune interactions. In inflamed tissue immune cell-derived opioid peptides activate opioid receptors on peripheral sensory nerves leading to potent analgesia. This is brought about by a release of opioids from inflammatory cells after stimulation by stress or corticotropin-releasing hormone (CRH). Immunocytes migrate from the circulation to inflamed tissue in multiple steps, including their rolling, adhesion, and transmigration through the vessel wall. This is orchestrated by adhesion molecules on leukocytes and vascular endothelium. Intercellular adhesion molecule-1 [ICAM-1 (or CD54)] is expressed by endothelium and mediates adhesion and extravasation of leukocytes. The goal of this study was to show that ICAM-1 regulates the homing of opioid-producing cells and the subsequent generation of analgesia within sites of painful inflammation. This was accomplished using immunofluorescence, flow cytometry, and behavioral (paw pressure) testing. We found that ICAM-1 is upregulated on the vascular endothelium, simultaneously with an enhanced immigration of opioid-containing immune cells into inflamed paw tissue. The intravenous administration of a monoclonal antibody against ICAM-1 markedly decreased the migration of opioid-containing leukocytes and of granulocytes, monocytes-macrophages, and T cells to the inflamed tissue. At the same time, circulating immunocytes increased in numbers, and macroscopic inflammation (hyperalgesia, paw volume, and paw temperature) remained primarily unchanged. Most importantly, peripheral opioid analgesia elicited either by cold water swim stress or by intraplantar administration of CRH was dramatically reduced. Together, these findings indicate that ICAM-1 expressed on vascular endothelium recruits immunocytes containing opioids to promote the local control of inflammatory pain.
Collapse
|
187
|
Steele AD, Szabo I, Bednar F, Rogers TJ. Interactions between opioid and chemokine receptors: heterologous desensitization. Cytokine Growth Factor Rev 2002; 13:209-22. [PMID: 12486875 DOI: 10.1016/s1359-6101(02)00007-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The opioid and chemokine receptors are both members of the seven transmembrane G protein-coupled receptor (GPCR) superfamily. Desensitization is believed to be a major element of the regulation of the function of these receptors, and recent findings suggest that both agonist-dependent (homologous) desensitization and heterologous desensitization can control receptor activity. The cross-desensitization between opioid and chemokine receptors has significant implications for our understanding of both the regulation of leukocyte trafficking, as well as the regulation of chemokine receptor function in inflammatory disease states. We also review findings which suggest that pro-inflammatory chemokine receptor-induced heterologous desensitization of opioid receptors has important implications for the regulation of opioid receptor function in the nervous system.
Collapse
Affiliation(s)
- Amber D Steele
- Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| | | | | | | |
Collapse
|
188
|
Abstract
The immune system and the nervous system are connected in a dynamic network that has important implications for psychology. First, if analyzed in functional terms, the immune system and the nervous system are not distinctly separated: The immune system can take part in neuronal signaling through the production of an array of transmitters and hormones, and brain cells can process and present antigen and produce immune proteins. Moreover, synapses seem to be formed between immune and neural cells in lymphoid tissue. The basis of this phenomenon may be a common evolutionary background of physiological systems that by tradition have been viewed as discrete rather than overlapping. Second, the immune system is actively involved in homeostatic regulation. Signals from immune cells can profoundly change the physiological state of the organism, with changes observed in metabolism, stress axes activity, behavior, motivation and cognition. Many of these changes have probably evolved to ease recuperation. Third, the activity in the immune system is dependent of homeostatic regulation by the neuroendocrine system in a biologically important network that is also capable of mediating psychological impact on immunity. In this review, it is argued that immunology should be ecological in nature and thereby related to psychological and neural science. Hypothetically, an ecological immunology will show cross-fertilizing properties, increasing the explanatory power of the seemingly disparate scientific disciplines involved.
Collapse
Affiliation(s)
- Mats Lekander
- Department of Clinical Neuroscience, Karolinska Institute, Sweden
| |
Collapse
|
189
|
Mousa SA, Machelska H, Schäfer M, Stein C. Immunohistochemical localization of endomorphin-1 and endomorphin-2 in immune cells and spinal cord in a model of inflammatory pain. J Neuroimmunol 2002; 126:5-15. [PMID: 12020952 DOI: 10.1016/s0165-5728(02)00049-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recently, two novel highly selective mu-opioid receptor (MOR) agonists, endomorphin-1 and endomorphin-2, have been isolated from bovine as well as human brains and were proposed to be the endogenous ligand for MOR. Later, endomorphin-1 and endomorphin-2 have been detected in the immune system of rats and humans using radioimmunoassay in combination with reverse-high-phase-liquid chromatography. In the present study, we analyzed the expression of endomorphin-1, endomorphin-2 and MOR by immunohistochemistry in a model of Freund's complete adjuvant (FCA)-induced painful inflammation. While MOR was upregulated on peripheral and central nerve terminals, inflammation did not alter endomorphin-2 expression in nerve fibers either in the dorsal horn of the spinal cord or in subcutaneous tissue. Endomorphin-1 and endomorphin-2 were expressed in immune cells (macrophage/monocytes) in the medullary region of the popliteal lymph nodes. The proportion of immunocytes (macrophage/monocytes, lymphocytes) containing endomorphin-1 and endomorphin-2 was increased in inflamed lymph nodes and subcutaneous paw tissue of animals with local inflammatory pain. Taken together, the upregulation of MOR and of its endogenous ligands endomorphin-1 and endomorphin-2 in immunocytes suggests an involvement of these opioid peptides in the peripheral control of inflammatory pain.
Collapse
Affiliation(s)
- Shaaban A Mousa
- Klinik für Anaesthesiologie und operative Intensivmedizin, Freie Universität Berlin, Universitätsklinikum Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany.
| | | | | | | |
Collapse
|
190
|
Abstract
Traditional acupuncture has been used for treating a variety of health conditions. In contrast, Western physicians limited acupuncture to the alleviation of pain. Concomitant with a recent view that not all kinds of pain are within the reach of acupuncture-induced relief, it has been suggested that some conditions other than pain can be effectively treated by this method. Increased release of the neuropeptide beta-endorphin was proposed to explain the antinociceptive function of acupuncture. Even if correct beta-endorphin cannot account for the effect of acupuncture in other conditions. Endorphins might be interacting with cytokines, some of which (e.g. interleukin-10) downregulate the inflammatory component of disorders in which acupuncture may be useful. We present a speculative notion of the view that acupuncture may amplify the interaction between neuropeptides and cytokines. A non-invasive approach, such as immune-committed cells harvested from blood of acupuncture-treated patients, could be used to examine this hypothesis. Inclusion of a placebo group might support the credibility of acupuncture.
Collapse
Affiliation(s)
- Ivan L Bonta
- Pharmacology, Erasmus University Rotterdam, The Netherlands.
| |
Collapse
|
191
|
Rossi A, Landolfo G, Sassi D, Franchini A, Ottaviani E. Immunocytochemical increased evidence of inducible nitric oxide synthase, tumor necrosis factor-alpha, and adrenocorticotropic hormone in human peritumoral lymph nodes. Appl Immunohistochem Mol Morphol 2002; 10:52-6. [PMID: 11893036 DOI: 10.1097/00129039-200203000-00009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In the current study, mesenteric and peritumoral lymph nodes surgically removed from patients with colon-rectum cancer were studied. Morphologic and immunocytochemical investigations demonstrated that mesenteric (control) and peritumoral lymph nodes of a same patient showed the same morphologic structure, but a different immunocytochemical pattern. Indeed, an increased immunoreactivity to anti-inducible nitric oxide synthase, anti-tumor necrosis factor-alpha, and anti-adrenocorticotropic hormone antibodies in the lymphatic tissue of peritumoral lymph nodes compared with mesenteric lymph nodes was observed. These findings suggest that in colon-rectum cancer, the pathologic event induces an increased expression of the molecules involved in the processes of inflammation and carcinogenesis that occurs earlier than the appearance of morphologic modifications.
Collapse
Affiliation(s)
- Aldo Rossi
- Surgical Clinic and Emergency Surgery, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | |
Collapse
|
192
|
CORRELATION OF ??-ENDORPHIN AND PROSTAGLANDIN E2 LEVELS IN PROSTATIC FLUID OF PATIENTS WITH CHRONIC PROSTATITIS WITH DIAGNOSIS AND TREATMENT RESPONSE. J Urol 2001. [DOI: 10.1097/00005392-200111000-00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
193
|
Correlation of beta-endorphin and prostaglandin E2 levels in prostatic fluid of patients with chronic prostatitis with diagnosis and treatment response. J Urol 2001. [PMID: 11586213 DOI: 10.1016/s0022-5347(05)65664-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE The chronic pelvic pain syndrome is a clinically defined symptom complex of unclear etiology. We have noted increased oxidative stress in the prostatic fluid of these patients, implying an active inflammatory response. Immune cells can produce the natural opioid beta-endorphin at the site of injury, which may modulate pain. We measured beta-endorphin and the inflammatory marker prostaglandin E2 in the expressed prostatic secretions of men with prostatitis, and correlated the results with symptoms. MATERIALS AND METHODS Expressed prostatic secretions samples from 70 patients and 8 asymptomatic controls were collected and frozen. beta-Endorphin and prostaglandin E2 were measured by enzyme-linked immunosorbent assay. Results were stratified according to prostatitis category and compared in individuals before and after therapy. RESULTS In symptomatic patients beta-endorphin and prostaglandin E2 were not significantly different in categories II, IIIa and IIIb expressed prostatic secretions but they were higher than in controls. The mean beta-endorphin level plus or minus standard error of mean in symptomatic patients was significantly higher (23.8 +/- 11 ng./ml. versus 8.7 +/- 4.7, p = 0.0001) and mean prostaglandin E2 was lower (6.01 +/- 2.9 ng./ml. versus 3.01 +/- 2.9, p = 0.001) after successful therapy with antibiotics or antioxidant phytotherapy, Prosta-Q (Farr Laboratories, Santa Clarita, California). CONCLUSIONS We observed a correlation of higher prostaglandin E2 and lower beta-endorphin in symptomatic men with chronic prostatitis. Increased oxidative stress and inflammation may induce prostaglandin E2 production that would inhibit beta-endorphin release. Treatment with therapeutic agents that decrease oxidative stress, such as antibiotics and antioxidant phytotherapy, may function at least partially by increasing beta-endorphin and decreasing prostaglandin E2.
Collapse
|
194
|
Abstract
Children frequently received no treatment, or inadequate treatment, for pain and for painful procedures. The newborn and critically ill children are especially vulnerable to no treatment or under-treatment. Nerve pathways essential for the transmission and perception of pain are present and functioning by 24 weeks of gestation. The failure to provide analgesia for pain results in rewiring the nerve pathways responsible for pain transmission in the dorsal horn of the spinal cord and results in increased pain perception for future painful results. Many children would withdraw or deny their pain in an attempt to avoid yet another terrifying and painful experiences, such as the intramuscular injections. Societal fears of opioid addiction and lack of advocacy are also causal factors in the under-treatment of pediatric pain. False beliefs about addictions and proper use of acetaminophen and other analgesics resulted in the failure to provide analgesia to children. All children even the newborn and critically ill require analgesia for pain and painful procedures. Unbelieved pain interferes with sleep, leads to fatigue and a sense of helplessness, and may result in increased morbidity or mortality.
Collapse
Affiliation(s)
- M Yaster
- Departments of Anesthesiology/Critical Care Medicine and Pediatrics, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | | |
Collapse
|
195
|
Mousa SA, Zhang Q, Sitte N, Ji R, Stein C. beta-Endorphin-containing memory-cells and mu-opioid receptors undergo transport to peripheral inflamed tissue. J Neuroimmunol 2001; 115:71-8. [PMID: 11282156 DOI: 10.1016/s0165-5728(01)00271-5] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Immunocyte-derived beta-endorphin can activate peripheral opioid receptors on sensory neurons to inhibit pain within inflamed tissue. This study examined mu-opioid receptors (MOR) on sensory nerves and beta-endorphin (END) in activated/memory CD4(+) cells (the predominant population homing to inflamed tissue). We found an upregulation of MOR in dorsal root ganglia, an increased axonal transport of MOR in the sciatic nerve and an accumulation of MOR in peripheral nerve terminals in Freund's adjuvant-induced hindpaw inflammation. A large number of CD4(+) cells containing beta-endorphin, but very few naive cells (CD45RC(+)), were observed in inflamed tissue, suggesting that this opioid is mainly present in activated/memory cells (CD4(+)/CD45RC(-)). Taken together, our results indicate an enhanced transport of both MOR and of the endogenous ligand beta-endorphin to injured tissue. This unique simultaneous upregulation of both receptors and ligands may serve to prevent excessive and/or chronic inflammatory pain.
Collapse
MESH Headings
- Animals
- Axonal Transport/immunology
- Blotting, Western
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Disease Models, Animal
- Freund's Adjuvant
- Ganglia, Spinal/cytology
- Ganglia, Spinal/metabolism
- Hindlimb
- Immunologic Memory/immunology
- Inflammation/chemically induced
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Ligation
- Male
- Neurons, Afferent/immunology
- Neurons, Afferent/metabolism
- Neurons, Afferent/pathology
- Rats
- Rats, Wistar
- Receptors, Opioid, mu/metabolism
- Sciatic Nerve/metabolism
- Sciatic Nerve/pathology
- Skin/immunology
- Skin/innervation
- Skin/pathology
- beta-Endorphin/metabolism
Collapse
Affiliation(s)
- S A Mousa
- Klinik für Anaesthesiologie und Operative Intensivmedizin, Freie Universität Berlin, Hindenburgdamm 30, D-12200, Berlin, Germany.
| | | | | | | | | |
Collapse
|
196
|
Abstract
1. Recent findings have suggested a significant involvement of the immune system in the control of pain. Immune cells contain opioid peptides that are released within inflamed tissue and act at opioid receptors on peripheral sensory nerve endings. It is also apparent that different types of lymphocytes contain beta-endorphin, memory T cells containing more beta-endorphin than naïve cells. 2. These findings highlight an integral link between immune cell migration and inflammatory pain. The present review highlights immune system involvement in the site-directed control of inflammatory pain. 3. Full-length mRNA transcripts for opioid precursor proteins are expressed in immune cells. Increased expression of pro-opiomelanocortin mRNA and beta-endorphin has been demonstrated in stimulated lymphocytes and lymphocytes from animals with inflammation. 4. Cytokines and corticotropin-releasing factor (CRF) release opioids from immune cells. Potent peripheral analgesia due to direct injection of CRF can be blocked by antagonists to CRF, antibodies to opioid peptides, antisense to CRF and opioid receptor-specific antagonists. The release of opioid peptides from lymphocytes is calcium dependent and opioid receptor specific. Furthermore, endogenous sources of opioid peptides produce potent analgesia when implanted into the spinal cord. 5. Activated immune cells migrate directly to inflamed tissue using cell adhesion molecules to adhere to the epithelial surface of the vasculature in inflamed tissue. Lymphocytes that have been activated can express opioid peptides. Memory type T cells that contain opioid peptides are present within inflamed tissue; naive cells are not present in inflamed tissue and do not contain opioid peptides. Inhibiting the migration of memory type T cells into inflamed tissue by blocking selectins results in reduced numbers of beta-endorphin-containing cells, a reduced quantity of beta-endorphin in inflamed paws and reduced stress- and CRF-induced peripheral analgesia. 6. Immunosuppression is associated with increased pain in patients. Moreover, immunosuppression results in decreased lymphocyte numbers as well as decreased analgesia in animal models.
Collapse
Affiliation(s)
- P J Cabot
- The School of Pharmacy, The University of Queensland, St Lucia, Australia.
| |
Collapse
|
197
|
Intraarticular Morphine and Bupivacaine Reduces Postoperative Pain After Rotator Cuff Repair. Reg Anesth Pain Med 2000. [DOI: 10.1097/00115550-200011000-00011] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
198
|
Brix-Christensen V, Goumon Y, Tønnesen E, Chew M, Bilfinger T, Stefano GB. Endogenous morphine is produced in response to cardiopulmonary bypass in neonatal pigs. Acta Anaesthesiol Scand 2000; 44:1204-8. [PMID: 11065199 DOI: 10.1034/j.1399-6576.2000.441004.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cardiopulmonary bypass (CPB) is associated with a systemic inflammatory response. Endogenous morphine production has previously been demonstrated in humans after cardiac surgery with CPB. It has been hypothesized that morphine plays a role as an anti-inflammatory mediator in the systemic inflammatory response. The aim of this study was to investigate if the CPB procedure in itself elicits an endogenous morphine production in neonatal pigs. METHODS Endogenous morphine production was measured in arterial blood in piglets exposed to sternotomy alone (sham group, n=10) or sternotomy and CPB (n=10). Blood samples were obtained immediately after the induction of anaesthesia, at the end of CPB and 4 h later. Morphine in arterial blood was detected by radioimmunoassay and confirmed by gas chromatography mass spectrometry. RESULTS Animals undergoing CPB showed detectable endogenous morphine concentrations immediately after CPB, with increased concentrations postoperatively. There was no measurable morphine production in the sham operated pigs. CONCLUSION The CPB procedures elicits an endogenous morphine production in neonatal pigs. This morphine response is analogous to the previously demonstrated response in patients subjected to cardiac surgery and CPB.
Collapse
Affiliation(s)
- V Brix-Christensen
- Department of Anaesthesiology and Intensive Care, Aarhus University Hospital, Denmark.
| | | | | | | | | | | |
Collapse
|
199
|
Stein C, Schäfer M, Machelska H. Why is morphine not the ultimate analgesic and what can be done to improve it? THE JOURNAL OF PAIN 2000; 1:51-6. [PMID: 14622843 DOI: 10.1054/jpai.2000.9820] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although opioids are unsurpassed in the treatment of acute and cancer pain, their use in chronic noncancer pain is clearly limited. This review discusses some open and controversial issues such as the balance between pain relief and side effects, whether all types of pain can be treated with opioids, and current efforts to develop opioids with an improved efficacy-side effect ratio. Whereas respiratory depression or tolerance are usually not major issues in long-term opioid use, it seems questionable whether opioids can produce an analgesic response in certain types of pain when there is a major affective component to the pain or when learned pain behavior is the main problem. Efforts to improve opioids have traditionally aimed at enhancing the selectivity of opioid receptor ligands towards mu-, delta-, and kappa-receptors. Another major strategy has been the search for opioid analgesics acting at opioid receptors outside the central nervous system, with the prospect to avoid centrally mediated side effects.
Collapse
Affiliation(s)
- C Stein
- Klinik für Aneaesthesiologie und operative Intensivmedizin, Universitätsklinikum Benjamin Franklin, Freie Universität Berlin, Germany.
| | | | | |
Collapse
|
200
|
Mousa SA, Machelska H, Schäfer M, Stein C. Co-expression of beta-endorphin with adhesion molecules in a model of inflammatory pain. J Neuroimmunol 2000; 108:160-70. [PMID: 10900350 DOI: 10.1016/s0165-5728(00)00284-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Opioid-containing immunocytes migrate to inflamed sites where they release beta-endorphin which activates peripheral opioid receptors and produces analgesia. The immigration of immunocytes to sites of inflammation is mediated by adhesion molecules. In this study, the expression of L-, P-, E-selectin and platelet-endothelial adhesion molecule-1 (PECAM-1) in relation to beta-endorphin expression was analyzed by immunohistochemistry in inflamed tissues. The proportion of immunocytes expressing L-selectin was increased in inflamed lymph nodes and subcutaneous paw tissue. P-selectin and PECAM-1 were constitutively expressed on endothelia of noninflamed lymph nodes and subcutaneous tissue and were upregulated in inflammation. beta-endorphin positive cells expressed L-selectin in lymph nodes and subcutaneous tissue. Upregulation of P-selectin and PECAM-1 and the co-localization of L-selectin and beta-endorphin in immunocytes suggest an important role of these adhesion molecules for the recruitment of immunocytes containing beta-endorphin to sites of painful inflammation.
Collapse
Affiliation(s)
- S A Mousa
- Klinik für Anaesthesiologie und operative Intensivmedizin, Freie Universität Berlin, Hindenburgdamm 30, D 12200, Berlin, Germany.
| | | | | | | |
Collapse
|