351
|
FACS analysis of neuronal-glial interactions in the nucleus accumbens following morphine administration. Psychopharmacology (Berl) 2013; 230:525-35. [PMID: 23793269 PMCID: PMC4134011 DOI: 10.1007/s00213-013-3180-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/04/2013] [Indexed: 10/26/2022]
Abstract
RATIONALE Glia, including astrocytes and microglia, can profoundly modulate neuronal function and behavior; however, very little is known about the signaling molecules that govern neuronal-glial communication and in turn affect behavior. Morphine treatment activates microglia and astrocytes in the nucleus accumbens (NAcc) to induce the synthesis of cytokines and chemokines, and this has important implications for addictive behavior. Blocking morphine-induced glial activation using the nonspecific glial inhibitor, ibudilast, has no effect on the initial rewarding properties of morphine, but completely prevents the relapse of drug-seeking behavior months later. OBJECTIVES We sought to determine the cellular source of these cytokines and chemokines in the NAcc in response to morphine, and the cell-type-specific expression pattern of their receptors to determine whether neurons have the capacity to respond to these immune signals directly. METHODS We used fluorescence-activated cell sorting of neurons (Thy1+), astrocytes (GLT1+), and microglia (CD11b+) from the NAcc for the analysis of cell type specific gene expression following morphine or saline treatment. RESULTS The results indicate that microglia and neurons each produce a subset of chemokines in response to morphine and that neurons have the capacity to respond directly to a select group of these chemokines via their receptors. In addition, we provide evidence that microglia are capable of responding directly to dopamine release in the NAcc. CONCLUSIONS Future studies will examine the mechanism(s) by which neurons respond to these immune signals produced by microglia in an effort to understand their effect on addictive behaviors.
Collapse
|
352
|
Abstract
This paper is the thirty-fifth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2012 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
353
|
Peri F, Calabrese V. Toll-like receptor 4 (TLR4) modulation by synthetic and natural compounds: an update. J Med Chem 2013; 57:3612-22. [PMID: 24188011 DOI: 10.1021/jm401006s] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Toll-like receptor 4 (TLR4), together with MD-2, binds bacterial endotoxins (E) with high affinity, triggering formation of the activated homodimer (E.MD-2.TLR4)2. Activated TLR4 induces intracellular signaling leading to activation of transcription factors that result in cytokine and chemokine production and initiation of inflammatory and immune responses. TLR4 also responds to endogenous ligands called danger associated molecular patterns (DAMPs). Increased sensitivity to infection and a variety of immune pathologies have been associated with either too little or too much TLR4 activation. We review here the molecular mechanisms of TLR4 activation (agonism) or inhibition (antagonism) by small organic molecules of both natural and synthetic origin. The role of co-receptors MD-2 and CD14 in the TLR4 modulation process is also discussed. Recent achievements in the field of chemical TLR4 modulation are reviewed, with special focus on nonclassical TLR4 ligands with a chemical structure different from that of lipid A.
Collapse
Affiliation(s)
- Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca , Piazza della Scienza, 2, 20126 Milano, Italy
| | | |
Collapse
|
354
|
Campbell LA, Avdoshina V, Rozzi S, Mocchetti I. CCL5 and cytokine expression in the rat brain: differential modulation by chronic morphine and morphine withdrawal. Brain Behav Immun 2013; 34:130-40. [PMID: 23968971 PMCID: PMC3795805 DOI: 10.1016/j.bbi.2013.08.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/12/2013] [Accepted: 08/13/2013] [Indexed: 01/23/2023] Open
Abstract
Opioids have been shown to influence the immune system and to promote the expression of pro-inflammatory cytokines in the central nervous system. However, recent data have shown that activation of opioid receptors increases the expression and release of the neuroprotective chemokine CCL5 from astrocytes in vitro. To further define the interaction between CCL5 and inflammation in response to opioids, we have examined the effect of chronic morphine and morphine withdrawal on the in vivo expression of CCL5 as well as of pro-inflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). Rats undergoing a chronic morphine paradigm (10 mg/kg increasing to 30 mg/kg, twice a day for 5 days) showed a twofold increase of CCL5 protein and mRNA within the cortex and striatum. No changes were observed in the levels of IL-1β and TNF-α. Naltrexone blocked the effect of morphine. A chronic morphine paradigm with no escalating doses (10 mg/kg, twice a day) did not alter CCL5 levels compared to saline-treated animals. On the contrary, rats undergoing spontaneous morphine withdrawal exhibited lower levels of CCL5 within the cortex as well as increased levels of pro-inflammatory cytokines and Iba-1 positive cells than saline-treated rats. Overall, these data suggest that morphine withdrawal may promote cytokines and other inflammatory responses that have the potential of exacerbating neuronal damage.
Collapse
Affiliation(s)
- Lee A. Campbell
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC 20057
| | - Valeriya Avdoshina
- Department of Neuroscience, Georgetown University Medical Center, Washington DC 20057
| | - Summer Rozzi
- Interdisciplinary Program of Neuroscience, Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC 20057
| | - Italo Mocchetti
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC 20057,Department of Neuroscience, Georgetown University Medical Center, Washington DC 20057
| |
Collapse
|
355
|
Harada S, Nakamoto K, Tokuyama S. The involvement of midbrain astrocyte in the development of morphine tolerance. Life Sci 2013; 93:573-8. [DOI: 10.1016/j.lfs.2013.08.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/02/2013] [Accepted: 08/13/2013] [Indexed: 01/28/2023]
|
356
|
Neri M, Panata L, Bacci M, Fiore C, Riezzo I, Turillazzi E, Fineschi V. Cytokines, chaperones and neuroinflammatory responses in heroin-related death: what can we learn from different patterns of cellular expression? Int J Mol Sci 2013; 14:19831-19845. [PMID: 24084728 PMCID: PMC3821589 DOI: 10.3390/ijms141019831] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 09/22/2013] [Accepted: 09/26/2013] [Indexed: 01/11/2023] Open
Abstract
Heroin (3,6-diacetylmorphine) has various effects on the central nervous system with several neuropathological alterations including hypoxic-ischemic brain damage from respiratory depressing effects and neuroinflammatory response. Both of these mechanisms induce the release of cytokines, chemokines and other inflammatory mediators by the activation of many cell types such as leucocytes and endothelial and glial cells, especially microglia, the predominant immunocompetent cell type within the central nervous system. The aim of this study is to clarify the correlation between intravenous heroin administration in heroin related death and the neuroinflammatory response. We selected 45 cases among autopsies executed for heroin-related death (358 total cases); immunohistochemical studies and Western blotting analyses were used to investigate the expression of brain markers such as tumor necrosis factor-α, oxygen-regulated protein 150, (interleukins) IL-1β, IL-6, IL-8, IL-10, IL-15, cyclooxygenase-2, heat shock protein 70, and CD68 (MAC387). Findings demonstrated that morphine induces inflammatory response and cytokine release. In particular, oxygen-regulated protein 150, cyclooxygenase-2, heat shock protein 70, IL-6 and IL-15 cytokines were over-expressed with different patterns of cellular expression.
Collapse
Affiliation(s)
- Margherita Neri
- Department of Forensic Pathology, University of Foggia, Ospedale Colonnello D’Avanzo, Viale degli Aviatori 1, Foggia 71100, Italy; E-Mails: (M.N.); (C.F.); (I.R.); (E.T.)
| | - Laura Panata
- Department of Forensic Pathology, University of Perugia, Via del Giochetto, Perugia 06100, Italy; E-Mails: (L.P.); (M.B.)
| | - Mauro Bacci
- Department of Forensic Pathology, University of Perugia, Via del Giochetto, Perugia 06100, Italy; E-Mails: (L.P.); (M.B.)
| | - Carmela Fiore
- Department of Forensic Pathology, University of Foggia, Ospedale Colonnello D’Avanzo, Viale degli Aviatori 1, Foggia 71100, Italy; E-Mails: (M.N.); (C.F.); (I.R.); (E.T.)
| | - Irene Riezzo
- Department of Forensic Pathology, University of Foggia, Ospedale Colonnello D’Avanzo, Viale degli Aviatori 1, Foggia 71100, Italy; E-Mails: (M.N.); (C.F.); (I.R.); (E.T.)
| | - Emanuela Turillazzi
- Department of Forensic Pathology, University of Foggia, Ospedale Colonnello D’Avanzo, Viale degli Aviatori 1, Foggia 71100, Italy; E-Mails: (M.N.); (C.F.); (I.R.); (E.T.)
| | - Vittorio Fineschi
- Department of Forensic Pathology, University of Foggia, Ospedale Colonnello D’Avanzo, Viale degli Aviatori 1, Foggia 71100, Italy; E-Mails: (M.N.); (C.F.); (I.R.); (E.T.)
| |
Collapse
|
357
|
Stevens CW, Aravind S, Das S, Davis RL. Pharmacological characterization of LPS and opioid interactions at the toll-like receptor 4. Br J Pharmacol 2013; 168:1421-9. [PMID: 23083095 DOI: 10.1111/bph.12028] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 10/03/2012] [Accepted: 10/10/2012] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Previous work in our laboratory showed opioid agents inhibit cytokine expression in astrocytes. Recently, Watkins and colleagues hypothesized that opioid agonists activate toll-like receptor 4 (TLR4) signalling, which leads to neuroinflammation. To test this hypothesis, we characterized LPS and opioid effects on TLR4 signalling in reporter cells. EXPERIMENTAL APPROACH NF-κB reporter cells expressing high levels of TLR4 were used to compare LPS and opioid effects on NF-κB activation, a pathway activated by TLR4 stimulation. KEY RESULTS LPS increased TLR4 signalling in a concentration-dependent manner and was antagonized by LPS antagonist (LPS-RS, from Rhodobacter sphaeroides). A concentration ratio analysis showed that LPS-RS was a competitive antagonist. The opioid agonists, morphine and fentanyl, produced minor activation of TLR4 signalling when given alone. When tested following LPS stimulation, opioid agonists inhibited NF-κB activation but this inhibition was not blocked by the general opioid antagonist, naloxone, nor by the selective μ opioid receptor antagonist, β-FNA. Indeed, both naloxone and β-FNA also inhibited NF-κB activation in reporter cells. Further examination of fentanyl and β-FNA effects revealed that both opioid agents inhibited LPS signalling in a non-competitive fashion. CONCLUSIONS AND IMPLICATIONS These results show that LPS-RS is a competitive antagonist at the TLR4 complex, and that both opioid agonists and antagonists inhibit LPS signalling in a non-competitive fashion through a non-GPCR, opioid site(s) in the TLR4 signalling pathway. If confirmed, existing opioid agents or other drug molecules more selective at this novel site may provide a new therapeutic approach to the treatment of neuroinflammation.
Collapse
Affiliation(s)
- C W Stevens
- Department of Pharmacology and Physiology, Oklahoma State University-Center for Health Sciences, Tulsa, OK 74107, USA.
| | | | | | | |
Collapse
|
358
|
Leonti M, Casu L. Traditional medicines and globalization: current and future perspectives in ethnopharmacology. Front Pharmacol 2013; 4:92. [PMID: 23898296 PMCID: PMC3722488 DOI: 10.3389/fphar.2013.00092] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/02/2013] [Indexed: 01/27/2023] Open
Abstract
The ethnopharmacological approach toward the understanding and appraisal of traditional and herbal medicines is characterized by the inclusions of the social as well as the natural sciences. Anthropological field-observations describing the local use of nature-derived medicines are the basis for ethnopharmacological enquiries. The multidisciplinary scientific validation of indigenous drugs is of relevance to modern societies at large and helps to sustain local health care practices. Especially with respect to therapies related to aging related, chronic and infectious diseases traditional medicines offer promising alternatives to biomedicine. Bioassays applied in ethnopharmacology represent the molecular characteristics and complexities of the disease or symptoms for which an indigenous drug is used in “traditional” medicine to variable depth and extent. One-dimensional in vitro approaches rarely cope with the complexity of human diseases and ignore the concept of polypharmacological synergies. The recent focus on holistic approaches and systems biology in medicinal plant research represents the trend toward the description and the understanding of complex multi-parameter systems. Ethnopharmacopoeias are non-static cultural constructs shaped by belief and knowledge systems. Intensified globalization and economic liberalism currently accelerates the interchange between local and global pharmacopoeias via international trade, television, the World Wide Web and print media. The increased infiltration of newly generated biomedical knowledge and introduction of “foreign” medicines into local pharmacopoeias leads to syncretic developments and generates a feedback loop. While modern and post-modern cultures and knowledge systems adapt and transform the global impact, they become more relevant for ethnopharmacology. Moreover, what is traditional, alternative or complementary medicine depends on the adopted historic-cultural perspective.
Collapse
Affiliation(s)
- Marco Leonti
- Department of Biomedical Sciences, University of Cagliari Cagliari, Italy
| | | |
Collapse
|
359
|
Weber ML, Chen C, Li Y, Farooqui M, Nguyen J, Poonawala T, Hebbel RP, Gupta K. Morphine stimulates platelet-derived growth factor receptor-β signalling in mesangial cells in vitro and transgenic sickle mouse kidney in vivo. Br J Anaesth 2013; 111:1004-12. [PMID: 23820675 DOI: 10.1093/bja/aet221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pain and renal dysfunction occur in sickle cell disease. Morphine used to treat pain also co-activates platelet-derived growth factor receptor-β (PDGFR-β), which can adversely affect renal disease. We examined the influence of morphine in mesangial cells in vitro and in mouse kidneys in vivo. METHODS > Mouse mesangial cells treated with 1 μM morphine in vitro or kidneys of transgenic homozygous or hemizygous sickle or control mice (n=3 for each), treated with morphine (0.75, 1.4, 2.14, 2.8, 3.6, and 4.3 mg kg(-1) day(-1) in two divided doses during the first, second, third, fourth, fifth, and sixth weeks, respectively), were used. Western blotting, bromylated deoxy uridine incorporation-based cell proliferation assay, reverse transcriptase-polymerase chain reaction, immunofluorescent microscopy, and blood/urine chemistry were used to analyse signalling, cell proliferation, opioid receptor (OP) expression, and renal function. RESULTS Morphine stimulated phosphorylation of PDGFR-β and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) to the same extent as induced by platelet-derived growth factor-BB (PDGF-BB) and promoted a two-fold increase in mesangial cell proliferation. The PDGFR-β inhibitor, AG1296, OP antagonists, and silencing of μ- and κ-OP abrogated morphine-induced MAPK/ERK phosphorylation and proliferation by ~100%. Morphine treatment of transgenic mice resulted in phosphorylation of PDGFR-β, MAPK/ERK, and signal transducer and activator of transcription 3 (Stat3) in the kidneys. Morphine inhibited micturition and blood urea nitrogen (BUN) clearance and increased BUN and urinary protein in sickle mice. CONCLUSION Morphine stimulates mitogenic signalling leading to mesangial cell proliferation and promotes renal dysfunction in sickle mice.
Collapse
Affiliation(s)
- M L Weber
- Division of Renal Diseases and Hypertension and
| | | | | | | | | | | | | | | |
Collapse
|
360
|
Weng Z, Lin Y, Zhang J, Yao S. Caspase inhibitors may attenuate opioid-induced hyperalgesia and tolerance via inhibiting microglial activation and neuroinflammation. JOURNAL OF MEDICAL HYPOTHESES AND IDEAS 2013. [DOI: 10.1016/j.jmhi.2012.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
361
|
Abstract
Sickle cell anemia (SCA) is an inherited disorder associated with severe lifelong pain and significant morbidity. The mechanisms of pain in SCA remain poorly understood. We show that mast cell activation/degranulation contributes to sickle pain pathophysiology by promoting neurogenic inflammation and nociceptor activation via the release of substance P in the skin and dorsal root ganglion. Mast cell inhibition with imatinib ameliorated cytokine release from skin biopsies and led to a correlative decrease in granulocyte-macrophage colony-stimulating factor and white blood cells in transgenic sickle mice. Targeting mast cells by genetic mutation or pharmacologic inhibition with imatinib ameliorates tonic hyperalgesia and prevents hypoxia/reoxygenation-induced hyperalgesia in sickle mice. Pretreatment with the mast cell stabilizer cromolyn sodium improved analgesia following low doses of morphine that were otherwise ineffective. Mast cell activation therefore underlies sickle pathophysiology leading to inflammation, vascular dysfunction, pain, and requirement for high doses of morphine. Pharmacological targeting of mast cells with imatinib may be a suitable approach to address pain and perhaps treat SCA.
Collapse
|
362
|
Shastri A, Bonifati DM, Kishore U. Innate immunity and neuroinflammation. Mediators Inflamm 2013; 2013:342931. [PMID: 23843682 PMCID: PMC3697414 DOI: 10.1155/2013/342931] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 05/15/2013] [Indexed: 01/07/2023] Open
Abstract
Inflammation of central nervous system (CNS) is usually associated with trauma and infection. Neuroinflammation occurs in close relation to trauma, infection, and neurodegenerative diseases. Low-level neuroinflammation is considered to have beneficial effects whereas chronic neuroinflammation can be harmful. Innate immune system consisting of pattern-recognition receptors, macrophages, and complement system plays a key role in CNS homeostasis following injury and infection. Here, we discuss how innate immune components can also contribute to neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Abhishek Shastri
- Centre for Infection, Immunity and Disease Mechanisms, Heinz Wolff Building, Brunel University, London UB8 3PH, UK
| | - Domenico Marco Bonifati
- Unit of Neurology, Department of Neurological Disorders, Santa Chiara Hospital, Largo Medaglie d'oro 1, 38100 Trento, Italy
| | - Uday Kishore
- Centre for Infection, Immunity and Disease Mechanisms, Heinz Wolff Building, Brunel University, London UB8 3PH, UK
| |
Collapse
|
363
|
Hutchinson MR, Watkins LR. Why is neuroimmunopharmacology crucial for the future of addiction research? Neuropharmacology 2013; 76 Pt B:218-27. [PMID: 23764149 DOI: 10.1016/j.neuropharm.2013.05.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/13/2013] [Accepted: 05/23/2013] [Indexed: 12/13/2022]
Abstract
A major development in drug addiction research in recent years has been the discovery that immune signaling within the central nervous system contributes significantly to mesolimbic dopamine reward signaling induced by drugs of abuse, and hence is involved in the presentation of reward behaviors. Additionally, in the case of opioids, these hypotheses have advanced through to the discovery of the novel site of opioid action at the innate immune pattern recognition receptor Toll-like receptor 4 as the necessary triggering event that engages this reward facilitating central immune signaling. Thus, the hypothesis of major proinflammatory contributions to drug abuse was born. This review will examine these key discoveries, but also address several key lingering questions of how central immune signaling is able to contribute in this fashion to the pharmacodynamics of drugs of abuse. It is hoped that by combining the collective wisdom of neuroscience, immunology and pharmacology, into Neuroimmunopharmacology, we may more fully understanding the neuronal and immune complexities of how drugs of abuse, such as opioids, create their rewarding and addiction states. Such discoveries will point us in the direction such that one day soon we might successfully intervene to successfully treat drug addiction. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Level 5, Medical School South, Frome Rd, Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
364
|
Lewis SS, Hutchinson MR, Zhang Y, Hund DK, Maier SF, Rice KC, Watkins LR. Glucuronic acid and the ethanol metabolite ethyl-glucuronide cause toll-like receptor 4 activation and enhanced pain. Brain Behav Immun 2013; 30:24-32. [PMID: 23348028 PMCID: PMC3641160 DOI: 10.1016/j.bbi.2013.01.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 01/09/2013] [Accepted: 01/10/2013] [Indexed: 12/19/2022] Open
Abstract
We have previously observed that the non-opioid morphine metabolite, morphine-3-glucuronide, enhances pain via a toll-like receptor 4 (TLR4) dependent mechanism. The present studies were undertaken to determine whether TLR4-dependent pain enhancement generalizes to other classes of glucuronide metabolites. In silico modeling predicted that glucuronic acid alone and ethyl glucuronide, a minor but long-lasting ethanol metabolite, would dock to the same MD-2 portion of the TLR4 receptor complex previously characterized as the docking site for morphine-3-glucuronide. Glucuronic acid, ethyl glucuronide and ethanol all caused an increase in TLR4-dependent reporter protein expression in a cell line transfected with TLR4 and associated co-signaling molecules. Glucuronic acid-, ethyl glucuronide-, and ethanol-induced increases in TLR4 signaling were blocked by the TLR4 antagonists LPS-RS and (+)-naloxone. Glucuronic acid and ethyl glucuronide both caused allodynia following intrathecal injection in rats, which was blocked by intrathecal co-administration of the TLR4 antagonist LPS-RS. The finding that ethyl glucuronide can cause TLR4-dependent pain could have implications for human conditions such as hangover headache and alcohol withdrawal hyperalgesia, as well as suggesting that other classes of glucuronide metabolites could have similar effects.
Collapse
Affiliation(s)
- Susannah S. Lewis
- Department of Psychology & Neuroscience, University of Colorado at Boulder, Boulder, Colorado, USA
| | - Mark R. Hutchinson
- Discipline of Pharmacology and Discipline of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Yingning Zhang
- Department of Psychology & Neuroscience, University of Colorado at Boulder, Boulder, Colorado, USA
| | - Dana K. Hund
- Department of Psychology & Neuroscience, University of Colorado at Boulder, Boulder, Colorado, USA
| | - Steven F. Maier
- Department of Psychology & Neuroscience, University of Colorado at Boulder, Boulder, Colorado, USA
| | - Kenner C. Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism National Institutes of Health, Rockville, Maryland, USA
| | - Linda R. Watkins
- Department of Psychology & Neuroscience, University of Colorado at Boulder, Boulder, Colorado, USA,Corresponding author: Linda R. Watkins, Department of Psychology, Campus Box 345, University of Colorado at Boulder, Boulder, Colorado, USA 80309-0345, , Fax: (303) 492-2967, Phone: (303) 492-7034
| |
Collapse
|
365
|
Theberge FR, Li X, Kambhampati S, Pickens CL, St Laurent R, Bossert JM, Baumann MH, Hutchinson MR, Rice KC, Watkins LR, Shaham Y. Effect of chronic delivery of the Toll-like receptor 4 antagonist (+)-naltrexone on incubation of heroin craving. Biol Psychiatry 2013; 73:729-37. [PMID: 23384483 PMCID: PMC3615146 DOI: 10.1016/j.biopsych.2012.12.019] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 12/10/2012] [Accepted: 12/27/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND Recent evidence implicates toll-like receptor 4 (TLR4) in opioid analgesia, tolerance, conditioned place preference, and self-administration. Here, we determined the effect of the TLR4 antagonist (+)-naltrexone (a μ-opioid receptor inactive isomer) on the time-dependent increases in cue-induced heroin seeking after withdrawal (incubation of heroin craving). METHODS In an initial experiment, we trained rats for 9 hours per day to self-administer heroin (.1 mg/kg/infusion) for 9 days; lever presses were paired with a 5-second tone-light cue. We then assessed cue-induced heroin seeking in 30-minute extinction sessions on withdrawal day 1; immediately after testing, we surgically implanted rats with Alzet minipumps delivering (+)-naltrexone (0, 7.5, 15, 30 mg/kg/day, subcutaneous) for 14 days. We then tested the rats for incubated cue-induced heroin seeking in 3-hour extinction tests on withdrawal day 13. RESULTS We found that chronic delivery of (+)-naltrexone via minipumps during the withdrawal phase decreased incubated cue-induced heroin seeking. In follow-up experiments, we found that acute injections of (+)-naltrexone immediately before withdrawal day 13 extinction tests had no effect on incubated cue-induced heroin seeking. Furthermore, chronic delivery of (+)-naltrexone (15 or 30 mg/kg/day) or acute systemic injections (15 or 30 mg/kg) had no effect on ongoing extended access heroin self-administration. Finally, in rats trained to self-administer methamphetamine (.1 mg/kg/infusion, 9 hours/day, 9 days), chronic delivery of (+)-naltrexone (30 mg/kg/day) during the withdrawal phase had no effect on incubated cue-induced methamphetamine seeking. CONCLUSIONS The present results suggest a critical role of TLR4 in the development of incubation of heroin, but not methamphetamine, craving.
Collapse
Affiliation(s)
- Florence R Theberge
- Intramural Research Program of the National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
366
|
Wang X, Grace PM, Pham MN, Cheng K, Strand KA, Smith C, Li J, Watkins LR, Yin H. Rifampin inhibits Toll-like receptor 4 signaling by targeting myeloid differentiation protein 2 and attenuates neuropathic pain. FASEB J 2013; 27:2713-22. [PMID: 23568774 DOI: 10.1096/fj.12-222992] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rifampin has been used for the treatment of bacterial infections for many years. Clinically, rifampin has been found to possess immunomodulatory effects. However, the molecular target responsible for the immunosuppressive effects of rifampin is not known. Herein, we show that rifampin binds to myeloid differentiation protein 2 (MD-2), the key coreceptor for innate immune TLR4. Rifampin blocked TLR4 signaling induced by LPS, including NF-κB activation and the overproduction of proinflammatory mediators nitric oxide, interleukin 1β, and tumor necrosis factor α in mouse microglia BV-2 cells and macrophage RAW 264.7 cells. Rifampin's inhibition of TLR4 signaling was also observed in immunocompetent rat primary macrophage, microglia, and astrocytes. Further, we show that rifampin (75 or 100 mg/kg b.i.d. for 3 d, intraperitoneal) suppressed allodynia induced by chronic constriction injury of the sciatic nerve and suppressed nerve injury-induced activation of microglia. Our findings indicate that MD-2 is a important target of rifampin in its inhibition of innate immune function and contributes to its clinically observed immune-suppressive effect. The results also suggest that rifampin may be repositioned as an agent for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, CO 80309, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
367
|
Doyle T, Esposito E, Bryant L, Cuzzocrea S, Salvemini D. NADPH-oxidase 2 activation promotes opioid-induced antinociceptive tolerance in mice. Neuroscience 2013; 241:1-9. [PMID: 23454539 DOI: 10.1016/j.neuroscience.2013.02.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Revised: 02/04/2013] [Accepted: 02/18/2013] [Indexed: 12/18/2022]
Abstract
The analgesic effectiveness of long-term opioid therapies is compromised by the development of antinociceptive tolerance linked to the overt production of peroxynitrite (ONOO(-), PN), the product of the interaction between superoxide (O2(-), SO) and nitric oxide (NO), and to neuroinflammatory processes. We have recently reported that in addition to post-translational nitration and inactivation of mitochondrial manganese superoxide dismutase (MnSOD), activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase holoenzyme (NOX) in the spinal cord is a major source for the overt production of superoxide-derived PN during the development of morphine-induced antinociceptive tolerance. However, the NOX complex involved in these processes is not known. The objective of these studies is to identify a potential role for the NOX2 complex, an enzyme involved in inflammation. Mice lacking the catalytic subunit of NOX2 (Nox2(-/-)) or its regulatory subunit, p47(phox) (p47(phox)(-/-)), developed antinociceptive tolerance similar to wildtype (wt) mice after 3 days of continuous morphine. However, while wt mice continue to develop tolerance by day six, morphine analgesia was restored in both Nox2(-/-) and p47(phox)(-/-) mice. Moreover, the loss of Nox2 or p47 did not affect acute morphine analgesia in naïve mice. In wt mice, antinociceptive tolerance was associated with increased activation of NOX, nitration of MnSOD, and proinflammatory cytokines production in the spinal cord. These events were markedly attenuated in Nox2(-/-) and p47(phox)(-/-) mice and instead, there was enhanced formation of antiinflammatory cytokine (IL4 and IL10) production. These results suggest that NOX2 activity provides a significant source of superoxide-derived PN to undertake post-translational modifications of mitochondrial MnSOD and to engage neuroinflammatory signaling in the spinal cord associated with opioid-induced antinociceptive tolerance. Thus, NOX2 may provide a potential target for adjuvant therapy to protect opioid analgesia.
Collapse
Affiliation(s)
- T Doyle
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St. Louis, MO 63104, USA
| | | | | | | | | |
Collapse
|
368
|
Little JW, Cuzzocrea S, Bryant L, Esposito E, Doyle T, Rausaria S, Neumann WL, Salvemini D. Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance. Pain 2013; 154:978-86. [PMID: 23590939 DOI: 10.1016/j.pain.2013.02.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/15/2013] [Accepted: 02/21/2013] [Indexed: 10/27/2022]
Abstract
Treatment of severe pain by morphine, the gold-standard opioid and a potent drug in our arsenal of analgesic medications, is limited by the eventual development of hyperalgesia and analgesic tolerance. We recently reported that systemic administration of a peroxynitrite (PN) decomposition catalyst (PNDC) or superoxide dismutase mimetic attenuates morphine hyperalgesia and antinociceptive tolerance and reduces PN-mediated mitochondrial nitroxidative stress in the spinal cord. These results suggest the potential involvement of spinal PN signaling in this setting; which was examined in the present study. PN removal with intrathecal delivery of manganese porphyrin-based dual-activity superoxide/PNDCs, MnTE-2-PyP(5+) and the more lipophilic MnTnHex-2-PyP(5+), blocked hyperalgesia and antinociceptive tolerance in rats. Noteworthy is that intrathecal MnTnHex-2-PyP(5+) prevented nitration and inactivation of mitochondrial manganese superoxide dismutase. Mitochondrial manganese superoxide dismutase inactivation enhances the superoxide-to-PN pathway by preventing the dismutation of superoxide to hydrogen peroxide, thus providing an important enzymatic source for PN formation. Additionally, intrathecal MnTnHex-2-PyP(5+) attenuated neuroimmune activation by preventing the activation of nuclear factor kappa B, extracellular-signal-regulated kinase and p38 mitogen activated protein kinases, and the enhanced levels of proinflammatory cytokines, interleukin (IL)-1β and IL-6, while increasing anti-inflammatory cytokines, IL-4 and IL-10. The role of PN was further confirmed using intrathecal or oral delivery of the superoxide-sparing PNDC, SRI-110. These results suggest that mitochondrial-derived PN triggers the activation of several biochemical pathways engaged in the development of neuroinflammation in the spinal cord that are critical to morphine hyperalgesia and tolerance, further supporting the potential of targeting PN as an adjunct to opiates to maintain pain relief.
Collapse
|
369
|
Ferrini F, Trang T, Mattioli TAM, Laffray S, Del’Guidice T, Lorenzo LE, Castonguay A, Doyon N, Zhang W, Godin AG, Mohr D, Beggs S, Vandal K, Beaulieu JM, Cahill C, Salter MW, De Koninck Y. Morphine hyperalgesia gated through microglia-mediated disruption of neuronal Cl⁻ homeostasis. Nat Neurosci 2013; 16:183-92. [PMID: 23292683 PMCID: PMC4974077 DOI: 10.1038/nn.3295] [Citation(s) in RCA: 340] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 12/05/2012] [Indexed: 01/18/2023]
Abstract
A major unresolved issue in treating pain is the paradoxical hyperalgesia produced by the gold-standard analgesic morphine and other opiates. We found that hyperalgesia-inducing treatment with morphine resulted in downregulation of the K(+)-Cl(-) co-transporter KCC2, impairing Cl(-) homeostasis in rat spinal lamina l neurons. Restoring the anion equilibrium potential reversed the morphine-induced hyperalgesia without affecting tolerance. The hyperalgesia was also reversed by ablating spinal microglia. Morphine hyperalgesia, but not tolerance, required μ opioid receptor-dependent expression of P2X4 receptors (P2X4Rs) in microglia and μ-independent gating of the release of brain-derived neurotrophic factor (BDNF) by P2X4Rs. Blocking BDNF-TrkB signaling preserved Cl(-) homeostasis and reversed the hyperalgesia. Gene-targeted mice in which Bdnf was deleted from microglia did not develop hyperalgesia to morphine. However, neither morphine antinociception nor tolerance was affected in these mice. Our findings dissociate morphine-induced hyperalgesia from tolerance and suggest the microglia-to-neuron P2X4-BDNF-KCC2 pathway as a therapeutic target for preventing hyperalgesia without affecting morphine analgesia.
Collapse
Affiliation(s)
- Francesco Ferrini
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
- Department of Veterinary Sciences, University of Turin, 10095, Grugliasco, Turin, Italy
| | - Tuan Trang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Departments of Comparative Biology & Experimental Medicine, and Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Theresa-Alexandra M. Mattioli
- Departments of Comparative Biology & Experimental Medicine, and Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sophie Laffray
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
| | - Thomas Del’Guidice
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
| | - Louis-Etienne Lorenzo
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
| | - Annie Castonguay
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
| | - Nicolas Doyon
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
| | - Wenbo Zhang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Antoine G. Godin
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
| | - Daniela Mohr
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Simon Beggs
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Karen Vandal
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
| | - Jean-Martin Beaulieu
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
| | - Catherine Cahill
- Department of Pharmacology & Toxicology, Queen’s University, Kingston, Ontario, Canada
| | - Michael W. Salter
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Yves De Koninck
- Institut Universitaire de santé mentale de Québec, Québec, G1J 2G3, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, Québec, G13 7P4, Canada
| |
Collapse
|
370
|
Snider SE, Hendrick ES, Beardsley PM. Glial cell modulators attenuate methamphetamine self-administration in the rat. Eur J Pharmacol 2013; 701:124-30. [PMID: 23375937 DOI: 10.1016/j.ejphar.2013.01.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 12/17/2012] [Accepted: 01/16/2013] [Indexed: 12/11/2022]
Abstract
Neuroinflammation induced by activated microglia and astrocytes can be elicited by drugs of abuse. Methamphetamine administration activates glial cells and increases proinflammatory cytokine production, and there is recent evidence of a linkage between glial cell activation and drug abuse-related behavior. We have previously reported that ibudilast (AV411; 3-isobutyryl-2-isopropylpyrazolo-[1,5-a]pyridine), which inhibits phosphodiesterase (PDE) and pro-inflammatory activity, blocks reinstatement of methamphetamine-maintained responding in rats, and that ibudilast and AV1013, an amino analog of ibudilast, which has similar glial-attenuating properties but limited PDE activity, attenuate methamphetamine-induced locomotor activity and sensitization in mice. The present study's objective was to determine whether co-administered ibudilast, AV1013, or minocycline, which is a tetracycline derivative that also suppresses methamphetamine-induced glial activation, would attenuate active methamphetamine i.v. self-administration in Long-Evans hooded rats. Rats were initially trained to press a lever for 0.1mg/kg/inf methamphetamine according to a FR1 schedule during 2-h daily sessions. Once stable responding was obtained, twice daily ibudilast (1, 7.5, 10mg/kg), AV1013 (1, 10, 30mg/kg), or once daily minocycline (10, 30, 60mg/kg), or their corresponding vehicles, were given i.p. for three consecutive days during methamphetamine (0.001, 0.03, 0.1mg/kg/inf) self-administration. Ibudilast, AV1013, and minocycline all significantly (p<0.05) reduced responding maintained by 0.03mg/kg/inf methamphetamine that had maintained the highest level of infusions under vehicle conditions. These results suggest that targeting glial cells may provide a novel approach to pharmacotherapy for treating methamphetamineabuse.
Collapse
Affiliation(s)
- Sarah E Snider
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | | | | |
Collapse
|
371
|
Schwarz JM, Bilbo SD. Adolescent morphine exposure affects long-term microglial function and later-life relapse liability in a model of addiction. J Neurosci 2013; 33:961-71. [PMID: 23325235 PMCID: PMC3713715 DOI: 10.1523/jneurosci.2516-12.2013] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 11/20/2012] [Accepted: 11/21/2012] [Indexed: 02/07/2023] Open
Abstract
Adolescence in humans represents a unique developmental time point associated with increased risk-taking behavior and experimentation with drugs of abuse. We hypothesized that exposure to drugs of abuse during adolescence may increase the risk of addiction in adulthood. To test this, rats were treated with a subchronic regimen of morphine or saline in adolescence, and their preference for morphine was examined using conditioned place preference (CPP) and drug-induced reinstatement in adulthood. The initial preference for morphine did not differ between groups; however, rats treated with morphine during adolescence showed robust reinstatement of morphine CPP after drug re-exposure in adulthood. This effect was not seen in rats pretreated with a subchronic regimen of morphine as adults, suggesting that exposure to morphine specifically during adolescence increases the risk of relapse to drug-seeking behavior in adulthood. We have previously established a role for microglia, the immune cells of the brain, and immune molecules in the risk of drug-induced reinstatement of morphine CPP. Thus, we examined the role of microglia within the nucleus accumbens of these rats and determined that rats exposed to morphine during adolescence had a significant increase in Toll-like receptor 4 (TLR4) mRNA and protein expression specifically on microglia. Morphine binds to TLR4 directly, and this increase in TLR4 was associated with exaggerated morphine-induced TLR4 signaling and microglial activation in rats previously exposed to morphine during adolescence. These data suggest that long-term changes in microglial function, caused by adolescent morphine exposure, alter the risk of drug-induced reinstatement in adulthood.
Collapse
Affiliation(s)
- Jaclyn M Schwarz
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina 27705, USA.
| | | |
Collapse
|
372
|
Swartjes M, Mooren RAG, Waxman AR, Arout C, van de Wetering K, den Hartigh J, Beijnen JH, Kest B, Dahan A. Morphine induces hyperalgesia without involvement of μ-opioid receptor or morphine-3-glucuronide. Mol Med 2012; 18:1320-6. [PMID: 23001479 DOI: 10.2119/molmed.2012.00244] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Accepted: 09/11/2012] [Indexed: 11/06/2022] Open
Abstract
Opioid-induced hyperalgesia (OIH) is a paradoxical increase in pain perception that may manifest during opioid treatment. For morphine, the metabolite morphine-3-glucuronide (M3G) is commonly believed to underlie this phenomenon. Here, in three separate studies, we empirically assess the role of M3G in morphine-induced hyperalgesia. In the first study, CD-1 mice injected with morphine (15 mg/kg subcutaneously) after pretreatment with the opioid receptor antagonist naltrexone (NTX) (15 mg/kg) showed tail withdrawal latency reductions indicative of hyperalgesia (2.5 ± 0.1 s at t = 30 min, P < 0.001 versus baseline). In these mice, the morphine/M3G concentration ratios versus effect showed a negative correlation (r(p) = -0.65, P < 0.001), indicating that higher morphine relative to M3G concentrations are associated with increased OIH. In the second study, similar hyperalgesic responses were observed in mice lacking the multidrug resistance protein 3 (MRP3) transporter protein (Mrp3(-/-) mice) in the liver and their wild-type controls (FVB mice; latency reductions: 3.1 ± 0.2 s at t = 30 min, P < 0.001 versus within-strain baseline). In the final study, the pharmacokinetics of morphine and M3G were measured in Mrp3(-/-) and FVB mice. Mrp3(-/-) mice displayed a significantly reduced capacity to export M3G into the systemic circulation, with plasma M3G concentrations just 7% of those observed in FVB controls. The data confirm previous literature that morphine causes hyperalgesia in the absence of opioid receptor activation but also indicate that this hyperalgesia may occur without a significant contribution of hepatic M3G. The relevance of these data to humans has yet to be demonstrated.
Collapse
Affiliation(s)
- Maarten Swartjes
- Department of Anesthesiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
373
|
Loram LC, Grace PM, Strand KA, Taylor FR, Ellis A, Berkelhammer D, Bowlin M, Skarda B, Maier SF, Watkins LR. Prior exposure to repeated morphine potentiates mechanical allodynia induced by peripheral inflammation and neuropathy. Brain Behav Immun 2012; 26:1256-64. [PMID: 22902523 PMCID: PMC3491069 DOI: 10.1016/j.bbi.2012.08.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 08/02/2012] [Accepted: 08/03/2012] [Indexed: 01/09/2023] Open
Abstract
Opioids, such as morphine, induce potent analgesia and are the gold standard for the treatment of acute pain. However, opioids also activate glia, inducing pro-inflammatory cytokine and chemokine production, which counter-regulates the analgesic properties of classical opioid receptor activation. It is not known how long these adverse pro-inflammatory effects last or whether prior morphine could sensitize the central nervous system (CNS) such that responses to a subsequent injury/inflammation would be exacerbated. Here, multiple models of inflammation or injury were induced two days after morphine (5mg/kg b.i.d., five days , s.c.) to test the generality of morphine sensitization of later pain. Prior repeated morphine potentiated the duration of allodynia from peripheral inflammatory challenges (complete Freund's adjuvant (CFA) into either hind paw skin or masseter muscle) and from peripheral neuropathy (mild chronic constriction injury (CCI) of the sciatic nerve). Spinal cord and trigeminal nucleus caudalis mRNAs were analyzed to identify whether repeated morphine was sufficient to alter CNS expression of pro-inflammatory response genes, measured two days after cessation of treatment. Prior morphine elevated IL-1β mRNA at both sites, MHC-II and TLR4 in the trigeminal nucleus caudalis but not spinal cord, but not glial activation markers at either site. Finally, in order to identify whether morphine sensitized pro-inflammatory cytokine release, spinal cord was isolated two days after morphine dosing for five days , and slices stimulated ex vivo with lipopolysaccharide. The morphine significantly induced TNFα protein release. Therefore, repeated morphine is able to sensitize subsequent CNS responses to immune challenges.
Collapse
Affiliation(s)
- Lisa C. Loram
- Corresponding author: Lisa Loram, PhD, Department of Psychology and Neuroscience, UCB 345, University of Colorado at Boulder, Boulder, Colorado, 80309 USA, , Fax: 303-492-2967, Phone: 720-224-2860
| | | | | | | | | | | | | | | | | | | |
Collapse
|
374
|
Repositioning antimicrobial agent pentamidine as a disruptor of the lateral interactions of transmembrane domain 5 of EBV latent membrane protein 1. PLoS One 2012; 7:e47703. [PMID: 23094078 PMCID: PMC3477141 DOI: 10.1371/journal.pone.0047703] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Accepted: 09/14/2012] [Indexed: 01/07/2023] Open
Abstract
The lateral transmembrane protein-protein interactions (PPI) have been regarded as "undruggable" despite their importance in many essential biological processes. The homo-trimerization of transmembrane domain 5 (TMD-5) of latent membrane protein 1 (LMP-1) is critical for the constitutive oncogenic activation of the Epstein-Barr virus (EBV). Herein we repurpose the antimicrobial agent pentamidine as a regulator of LMP-1 TMD-5 lateral interactions. The results of ToxR assay, tryptophan fluorescence assay, courmarin fluorescence dequenching assay, and Bis-Tris sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) consistently show pentamidine disrupts LMP-1 TMD-5 lateral interactions. Furthermore, pentamidine inhibits LMP-1 signaling, inducing cellular apoptosis and suppressing cell proliferation in the EBV infected B cells. In contrast, EBV negative cells are less susceptible to pentamidine. This study provides a novel non-peptide small molecule agent for regulating LMP-1 TMD-5 lateral interactions.
Collapse
|
375
|
Kwok YH, Hutchinson MR, Gentgall MG, Rolan PE. Increased responsiveness of peripheral blood mononuclear cells to in vitro TLR 2, 4 and 7 ligand stimulation in chronic pain patients. PLoS One 2012; 7:e44232. [PMID: 22937165 PMCID: PMC3429430 DOI: 10.1371/journal.pone.0044232] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 07/30/2012] [Indexed: 01/23/2023] Open
Abstract
Glial activation via Toll-like receptor (TLR) signaling has been shown in animals to play an important role in the initiation and establishment of chronic pain. However, our ability to assess this central immune reactivity in clinical pain populations is currently lacking. Peripheral blood mononuclear cells (PBMCs) are an accessible source of TLR expressing cells that may mirror similarities in TLR responsiveness of the central nervous system. The aim of this study was to characterize the IL-1β response to various TLR agonists in isolated PBMCs from chronic pain sufferers (on and not on opioids) and pain-free controls. Venous blood was collected from 11 chronic pain sufferers on opioids (≥ 20 mg of morphine / day), 8 chronic pain sufferers not on opioids and 11 pain-free controls. PBMCs were isolated and stimulated in vitro with a TLR2 (Pam3CSK4), TLR4 (LPS) or TLR7 (imiquimod) agonist. IL-1β released into the supernatant was measured with ELISA. Significantly increased IL-1β expression was found in PBMCs from chronic pain sufferers (on and not on opioids) compared with pain-free controls for TLR2 (F((6, 277)) = 15, P<0.0001), TLR4 (F((8, 263)) = 3, P = 0.002) and TLR7 (F((2,201)) = 5, P = 0.005) agonists. These data demonstrate that PBMCs from chronic pain sufferers were more responsive to TLR agonists compared with controls, suggesting peripheral cells may have the potential to become a source of biomarkers for chronic pain.
Collapse
Affiliation(s)
- Yuen H Kwok
- Discipline of Pharmacology, School of Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | | | | | | |
Collapse
|
376
|
Due MR, Piekarz AD, Wilson N, Feldman P, Ripsch MS, Chavez S, Yin H, Khanna R, White FA. Neuroexcitatory effects of morphine-3-glucuronide are dependent on Toll-like receptor 4 signaling. J Neuroinflammation 2012; 9:200. [PMID: 22898544 PMCID: PMC3519737 DOI: 10.1186/1742-2094-9-200] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 07/31/2012] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Multiple adverse events are associated with the use of morphine for the treatment of chronic non-cancer pain, including opioid-induced hyperalgesia (OIH). Mechanisms of OIH are independent of opioid tolerance and may involve the morphine metabolite morphine-3-glucuronide (M3G). M3G exhibits limited affinity for opioid receptors and no analgesic effect. Previous reports suggest that M3G can act via the Toll-like receptor 4 (TLR4)/myeloid differentiation protein-2 (MD-2) heterodimer in the central nervous system to elicit pain. METHODS Immunoblot and immunocytochemistry methods were used to characterize the protein expression of TLR4 present in lumbar dorsal root ganglion (DRG). Using in vitro intracellular calcium and current clamp techniques, we determined whether TLR4 activation as elicited by the prototypical agonists of TLR4, lipopolysaccharide (LPS) and M3G, contributed to changes in intracellular calcium and increased excitation. Rodents were also injected with M3G to determine the degree to which M3G-induced tactile hyperalgesia could be diminished using either a small molecule inhibitor of the MD-2/TLR4 complex in rats or TLR4 knockout mice. Whole cell voltage-clamp recordings were made from small- and medium-diameter DRG neurons (25 μm < DRG diameter <45 μm) for both control and M3G-treated neurons to determine the potential influence on voltage-gated sodium channels (NaVs). RESULTS We observed that TLR4 immunoreactivity was present in peptidergic and non-peptidergic sensory neurons in the DRG. Non-neuronal cells in the DRG lacked evidence of TLR4 expression. Approximately 15% of assayed small- and medium-diameter sensory neurons exhibited a change in intracellular calcium following LPS administration. Both nociceptive and non-nociceptive neurons were observed to respond, and approximately 40% of these cells were capsaicin-insensitive. Increased excitability observed in sensory neurons following LPS or M3G could be eliminated using Compound 15, a small molecule inhibitor of the TLR4/MD-2 complex. Likewise, systemic injection of M3G induced rapid tactile, but not thermal, nociceptive behavioral changes in the rat, which were prevented by pre-treating animals with Compound 15. Unlike TLR4 wild-type mice, TLR4 knockout mice did not exhibit M3G-induced hyperalgesia. As abnormal pain sensitivity is often associated with NaVs, we predicted that M3G acting via the MD-2/TLR4 complex may affect the density and gating of NaVs in sensory neurons. We show that M3G increases tetrodotoxin-sensitive and tetrodotoxin-resistant (NaV1.9) current densities. CONCLUSIONS These outcomes provide evidence that M3G may play a role in OIH via the TLR4/MD-2 heterodimer complex and biophysical properties of tetrodotoxin-sensitive and tetrodotoxin-resistant NaV currents.
Collapse
Affiliation(s)
- Michael R Due
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
377
|
Hutchinson MR, Northcutt AL, Hiranita T, Wang X, Lewis SS, Thomas J, van Steeg K, Kopajtic TA, Loram LC, Sfregola C, Galer E, Miles NE, Bland ST, Amat J, Rozeske RR, Maslanik T, Chapman TR, Strand KA, Fleshner M, Bachtell RK, Somogyi AA, Yin H, Katz JL, Rice KC, Maier SF, Watkins LR. Opioid activation of toll-like receptor 4 contributes to drug reinforcement. J Neurosci 2012; 32:11187-200. [PMID: 22895704 PMCID: PMC3454463 DOI: 10.1523/jneurosci.0684-12.2012] [Citation(s) in RCA: 231] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 06/14/2012] [Accepted: 06/25/2012] [Indexed: 01/08/2023] Open
Abstract
Opioid action was thought to exert reinforcing effects solely via the initial agonism of opioid receptors. Here, we present evidence for an additional novel contributor to opioid reward: the innate immune pattern-recognition receptor, toll-like receptor 4 (TLR4), and its MyD88-dependent signaling. Blockade of TLR4/MD2 by administration of the nonopioid, unnatural isomer of naloxone, (+)-naloxone (rats), or two independent genetic knock-outs of MyD88-TLR4-dependent signaling (mice), suppressed opioid-induced conditioned place preference. (+)-Naloxone also reduced opioid (remifentanil) self-administration (rats), another commonly used behavioral measure of drug reward. Moreover, pharmacological blockade of morphine-TLR4/MD2 activity potently reduced morphine-induced elevations of extracellular dopamine in rat nucleus accumbens, a region critical for opioid reinforcement. Importantly, opioid-TLR4 actions are not a unidirectional influence on opioid pharmacodynamics, since TLR4(-/-) mice had reduced oxycodone-induced p38 and JNK phosphorylation, while displaying potentiated analgesia. Similar to our recent reports of morphine-TLR4/MD2 binding, here we provide a combination of in silico and biophysical data to support (+)-naloxone and remifentanil binding to TLR4/MD2. Collectively, these data indicate that the actions of opioids at classical opioid receptors, together with their newly identified TLR4/MD2 actions, affect the mesolimbic dopamine system that amplifies opioid-induced elevations in extracellular dopamine levels, therefore possibly explaining altered opioid reward behaviors. Thus, the discovery of TLR4/MD2 recognition of opioids as foreign xenobiotic substances adds to the existing hypothesized neuronal reinforcement mechanisms, identifies a new drug target in TLR4/MD2 for the treatment of addictions, and provides further evidence supporting a role for central proinflammatory immune signaling in drug reward.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/blood
- Analysis of Variance
- Animals
- Conditioning, Operant/drug effects
- Conditioning, Operant/physiology
- Dopamine/metabolism
- Dose-Response Relationship, Drug
- Drug Administration Routes
- Hyperalgesia/drug therapy
- Hyperalgesia/physiopathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Microdialysis
- Mitogen-Activated Protein Kinase 1/metabolism
- Models, Molecular
- Myeloid Differentiation Factor 88/deficiency
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/metabolism
- Pain Threshold/drug effects
- Pain Threshold/physiology
- Phosphorylation/drug effects
- Protein Binding/drug effects
- Protein Binding/genetics
- Rats
- Rats, Sprague-Dawley
- Reaction Time/drug effects
- Reinforcement, Psychology
- Self Administration
- Signal Transduction/drug effects
- Time Factors
- Toll-Like Receptor 4/agonists
- Toll-Like Receptor 4/deficiency
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- M R Hutchinson
- Department of Psychology and Neuroscience, University of Colorado-Boulder, Boulder, Colorado 80309, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
378
|
Ouyang H, Liu S, Zeng W, Levitt RC, Candiotti KA, Hao S. An emerging new paradigm in opioid withdrawal: a critical role for glia-neuron signaling in the periaqueductal gray. ScientificWorldJournal 2012; 2012:940613. [PMID: 22919361 PMCID: PMC3419410 DOI: 10.1100/2012/940613] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 06/06/2012] [Indexed: 11/25/2022] Open
Abstract
The chronic use of opiates (i.e., narcotics such as the natural derivatives of opium including morphine or codeine) or opioids (i.e., semisynthetic derivatives of opium and other molecules that activate opioid receptors) induces dependence, which is associated with various specific behavioral and somatic signs after their withdrawal or after the administration of an opioid antagonist. Among the brain regions implicated in opiate dependence and withdrawal, the periaqueductal gray area (PAG) appears to be critical in regulating the complex signs and symptoms of opioid withdrawal. Numerous neurochemical mechanisms in the PAG have been identified that may contribute to the opioid withdrawal syndrome. Accumulating evidence suggests that glial activation leading to the release of proinflammatory molecules acting on neurons is important in the complex syndrome of opioid dependence and withdrawal. This paper focuses on the recent advances in our understanding of the vital role that glia-neuron interactions play in opioid dependence and withdrawal within the PAG. We summarize those neurochemical mechanisms associated with opioid withdrawal including the recently defined importance of TNFα release from activated glial cells that communicate with TNF receptors on PAG neurons.
Collapse
Affiliation(s)
- Handong Ouyang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
379
|
Kovács KJ. Microglia and drug-induced plasticity in reward-related neuronal circuits. Front Mol Neurosci 2012; 5:74. [PMID: 22707932 PMCID: PMC3374147 DOI: 10.3389/fnmol.2012.00074] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 05/25/2012] [Indexed: 12/16/2022] Open
Affiliation(s)
- Krisztina J Kovács
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine Budapest, Hungary
| |
Collapse
|
380
|
Reece AS. Epidemiologic and Molecular Pathophysiology of Chronic Opioid Dependence and the Place of Naltrexone Extended-Release Formulations in its Clinical Management. Subst Abuse 2012; 6:115-33. [PMID: 23055738 PMCID: PMC3465087 DOI: 10.4137/sart.s9031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Naltrexone implants and depot injections (NI) are a novel form of treatment for opiate dependence (OD). Major questions relate to their absolute and relative efficacy and safety. Opportunely, six recent clinical trial data from several continents have uniformly provided dramatic evidence of the potent, dose-related and highly significant efficacy of NI, with minimal or manageable accompanying toxicity and safety concerns. The opiate-free lifestyle is attained significantly more often with NI adjusted O.R. = 6.00 (95% C.I. 3.86–9.50), P < 10−10. Other drug use and drug craving are also rapidly reduced. The optimum manner in which to commence NI remains to be established. Of particular relevance is the relative safety of NI compared to the chronic opiate agonists (COA) usually employed, as the long-term toxicity of COA is only just being elucidated. Large population-based studies have found elevated rates of cardiovascular disease, six cancers, liver and respiratory disease, and all-cause mortality in COA. Whilst opiates have been shown to trigger numerous molecular pathways, the most interesting is the demonstration that the opiate morphinan’s nucleus binds to the endotoxin groove of the TLR4-MD2 heterodimer. This has the effect of triggering a low grade endotoxaemic-like state, which over time may account for these protean clinical findings, an effect which is reversed by opiate antagonists. This emerging evidence suggests an exciting new treatment paradigm for OD and a corresponding increase in the role of NI in treatment.
Collapse
Affiliation(s)
- Albert Stuart Reece
- School of Psychiatry and Clinical Neurosciences, University of Western Australia
| |
Collapse
|