1
|
Furdui A, da Silveira Scarpellini C, Montandon G. Mu-opioid receptors in tachykinin-1-positive cells mediate the respiratory and antinociceptive effects of the opioid fentanyl. Br J Pharmacol 2024. [PMID: 39506356 DOI: 10.1111/bph.17369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/26/2024] [Accepted: 09/03/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND AND PURPOSE Opioid drugs are potent analgesics that carry the risk of respiratory side effects due to actions on μ-opioid receptors (MORs) in brainstem regions that control respiration. Substance P is encoded by the Tac1 gene and is expressed in neurons regulating breathing, nociception, and locomotion. Tac1-positive cells also express MORs in brainstem regions mediating opioid-induced respiratory depression. We determined the role of Tac1-positive cells in mediating the respiratory effects of opioid drugs. EXPERIMENTAL APPROACH In situ hybridization was used to determine Oprm1 mRNA expression (gene encoding MORs) in Tac1-positive cells in regions regulating respiratory depression by opioid drugs. Conditional knockout mice lacking functional MORs in Tac1-positive cells were produced and the respiratory and locomotor responses to the opioid analgesic fentanyl were assessed using whole-body plethysmography. A tail immersion assay was used to assess the antinociceptive response to fentanyl. KEY RESULTS Oprm1 mRNA was highly expressed (>80%) in subpopulations of Tac1-positive cells in the preBötzinger Complex, nucleus tractus solitarius, and Kölliker-Fuse/lateral parabrachial region. Conditionally knocking out MORs in Tac1-positive cells abolished the effects of fentanyl on respiratory rate, relative tidal volume, and relative minute ventilation compared with control mice. Importantly, the antinociceptive response of fentanyl was eliminated in mice lacking functional MORs in Tac1-positive cells, whereas locomotor effects induced by fentanyl were preserved. CONCLUSIONS AND IMPLICATIONS Our findings suggest that Tac1-positive cells mediate the respiratory depressive and antinociceptive effects of the opioid fentanyl, providing important insights for the development of pain therapies with reduced risk of respiratory side effects.
Collapse
Affiliation(s)
- Andreea Furdui
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Gaspard Montandon
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Ochandarena NE, Niehaus JK, Tassou A, Scherrer G. Cell-type specific molecular architecture for mu opioid receptor function in pain and addiction circuits. Neuropharmacology 2023; 238:109597. [PMID: 37271281 PMCID: PMC10494323 DOI: 10.1016/j.neuropharm.2023.109597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/13/2023] [Indexed: 06/06/2023]
Abstract
Opioids are potent analgesics broadly used for pain management; however, they can produce dangerous side effects including addiction and respiratory depression. These harmful effects have led to an epidemic of opioid abuse and overdose deaths, creating an urgent need for the development of both safer pain medications and treatments for opioid use disorders. Both the analgesic and addictive properties of opioids are mediated by the mu opioid receptor (MOR), making resolution of the cell types and neural circuits responsible for each of the effects of opioids a critical research goal. Single-cell RNA sequencing (scRNA-seq) technology is enabling the identification of MOR-expressing cell types throughout the nervous system, creating new opportunities for mapping distinct opioid effects onto newly discovered cell types. Here, we describe molecularly defined MOR-expressing neuronal cell types throughout the peripheral and central nervous systems and their potential contributions to opioid analgesia and addiction.
Collapse
Affiliation(s)
- Nicole E Ochandarena
- Neuroscience Curriculum, Biological and Biomedical Sciences Program, The University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Jesse K Niehaus
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
3
|
Rayala R, Tiller A, Majumder SA, Stacy HM, Eans SO, Nedovic A, McLaughlin JP, Cudic P. Solid-Phase Synthesis of the Bicyclic Peptide OL-CTOP Containing Two Disulfide Bridges, and an Assessment of Its In Vivo μ-Opioid Receptor Antagonism after Nasal Administration. Molecules 2023; 28:1822. [PMID: 36838810 PMCID: PMC9963138 DOI: 10.3390/molecules28041822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/02/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
New strategies facilitate the design of cyclic peptides which can penetrate the brain. We have designed a bicyclic peptide, OL-CTOP, composed of the sequences of a selective μ-opioid receptor antagonist, CTOP (f-cyclo(CYwOTX)T) (X = penicillamine, Pen; O = ornithine) and odorranalectin, OL (YASPK-cyclo(CFRYPNGVLAC)T), optimized its solid-phase synthesis and demonstrated its ability for nose-to-brain delivery and in vivo activity. The differences in reactivity of Cys and Pen thiol groups protected with trityl and/or acetamidomethyl protecting groups toward I2 in different solvents were exploited for selective disulfide bond formation on the solid phase. Both the single step and the sequential strategy applied to macrocyclization reactions generated the desired OL-CTOP, with the sequential strategy yielding a large quantity and better purity of crude OL-CTOP. Importantly, intranasally (i.n.s.) administered OL-CTOP dose-dependently antagonized the analgesic effect of morphine administered to mice through the intracerebroventricular route and prevented morphine-induced respiratory depression. In summary, the results demonstrate the feasibility of our solid-phase synthetic strategy for the preparation of the OL-CTOP bicyclic peptide containing two disulfide bonds and reveal the potential of odorranalectin for further modifications and the targeted delivery to the brain.
Collapse
Affiliation(s)
- Ramanjaneyulu Rayala
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| | - Annika Tiller
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| | - Shahayra A. Majumder
- Department of Pharmacodynamics, School of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, FL 32610, USA
| | - Heather M. Stacy
- Department of Pharmacodynamics, School of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, FL 32610, USA
| | - Shainnel O. Eans
- Department of Pharmacodynamics, School of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, FL 32610, USA
| | - Aleksandra Nedovic
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, School of Pharmacy, University of Florida, 1345 Center Drive, Gainesville, FL 32610, USA
| | - Predrag Cudic
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| |
Collapse
|
4
|
The Opioid System in Depression. Neurosci Biobehav Rev 2022; 140:104800. [PMID: 35914624 PMCID: PMC10166717 DOI: 10.1016/j.neubiorev.2022.104800] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 12/16/2022]
Abstract
Opioid receptors are widely distributed throughout the brain and play an essential role in modulating aspects of human mood, reward, and well-being. Accumulating evidence indicates the endogenous opioid system is dysregulated in depression and that pharmacological modulators of mu, delta, and kappa opioid receptors hold potential for the treatment of depression. Here we review animal and clinical data, highlighting evidence to support: dysregulation of the opioid system in depression, evidence for opioidergic modulation of behavioural processes and brain regions associated with depression, and evidence for opioidergic modulation in antidepressant responses. We evaluate clinical trials that have examined the safety and efficacy of opioidergic agents in depression and consider how the opioid system may be involved in the effects of other treatments, including ketamine, that are currently understood to exert antidepressant effects through non-opioidergic actions. Finally, we explore key neurochemical and molecular mechanisms underlying the potential therapeutic effects of opioid system engagement, that together provides a rationale for further investigation into this relevant target in the treatment of depression.
Collapse
|
5
|
Hsieh YL, Yang CC, Yang NP. Ultra-Low Frequency Transcutaneous Electrical Nerve Stimulation on Pain Modulation in a Rat Model with Myogenous Temporomandibular Dysfunction. Int J Mol Sci 2021; 22:ijms22189906. [PMID: 34576074 PMCID: PMC8465049 DOI: 10.3390/ijms22189906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 12/22/2022] Open
Abstract
Masticatory myofascial pain (MMP) is one of the most common causes of chronic orofacial pain in patients with temporomandibular disorders. To explore the antinociceptive effects of ultra-low frequency transcutaneous electrical nerve stimulation (ULF-TENS) on alterations of pain-related biochemicals, electrophysiology and jaw-opening movement in an animal model with MMP, a total of 40 rats were randomly and equally assigned to four groups; i.e., animals with MMP receiving either ULF-TENS or sham treatment, as well as those with sham-MMP receiving either ULF-TENS or sham treatment. MMP was induced by electrically stimulated repetitive tetanic contraction of masticatory muscle for 14 days. ULF-TENS was then performed at myofascial trigger points of masticatory muscles for seven days. Measurable outcomes included maximum jaw-opening distance, prevalence of endplate noise (EPN), and immunohistochemistry for substance P (SP) and μ-opiate receptors (MOR) in parabrachial nucleus and c-Fos in rostral ventromedial medulla. There were significant improvements in maximum jaw-opening distance and EPN prevalence after ULF-TENS in animals with MMP. ULF-TENS also significantly reduced SP overexpression, increased MOR expression in parabrachial nucleus, and increased c-Fos expression in rostral ventromedial medulla. ULF-TENS may represent a novel and applicable therapeutic approach for improvement of orofacial pain induced by MMP.
Collapse
Affiliation(s)
- Yueh-Ling Hsieh
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung 406040, Taiwan;
- Correspondence: ; Tel.: +886-4-22053366 (ext. 7312)
| | - Chen-Chia Yang
- Kao-An Physical Medicine and Rehabilitation Clinic, Taichung 406040, Taiwan;
| | - Nian-Pu Yang
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung 406040, Taiwan;
| |
Collapse
|
6
|
Cuitavi J, Hipólito L, Canals M. The Life Cycle of the Mu-Opioid Receptor. Trends Biochem Sci 2021; 46:315-328. [PMID: 33127216 DOI: 10.1016/j.tibs.2020.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Opioid receptors (ORs) are undisputed targets for the treatment of pain. Unfortunately, targeting these receptors therapeutically poses significant challenges including addiction, dependence, tolerance, and the appearance of side effects, such as respiratory depression and constipation. Moreover, misuse of prescription and illicit narcotics has resulted in the current opioid crisis. The mu-opioid receptor (MOR) is the cellular mediator of the effects of most commonly used opioids, and is a prototypical G protein-coupled receptor (GPCR) where new pharmacological, signalling and cell biology concepts have been coined. This review summarises the knowledge of the life cycle of this therapeutic target, including its biogenesis, trafficking to and from the plasma membrane, and how the regulation of these processes impacts its function and is related to pathophysiological conditions.
Collapse
Affiliation(s)
- Javier Cuitavi
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of València, Burjassot, Spain
| | - Lucía Hipólito
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of València, Burjassot, Spain
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, the Midlands, UK.
| |
Collapse
|
7
|
Abstract
Hybrid compounds (also known as chimeras, designed multiple ligands, bivalent compounds) are chemical units where two active components, usually possessing affinity and selectivity for distinct molecular targets, are combined as a single chemical entity. The rationale for using a chimeric approach is well documented as such novel drugs are characterized by their enhanced enzymatic stability and biological activity. This allows their use at lower concentrations, increasing their safety profile, particularly when considering undesirable side effects. In the group of synthetic bivalent compounds, drugs combining pharmacophores having affinities toward opioid and neurokinin-1 receptors have been extensively studied as potential analgesic drugs. Indeed, substance P is known as a major endogenous modulator of nociception both in the peripheral and central nervous systems. Hence, synthetic peptide fragments showing either agonism or antagonism at neurokinin 1 receptor were both assigned with analgesic properties. However, even though preclinical studies designated neurokinin-1 receptor antagonists as promising analgesics, early clinical studies revealed a lack of efficacy in human. Nevertheless, their molecular combination with enkephalin/endomorphin fragments has been considered as a valuable approach to design putatively promising ligands for the treatment of pain. This paper is aimed at summarizing a 20-year journey to the development of potent analgesic hybrid compounds involving an opioid pharmacophore and devoid of unwanted side effects. Additionally, the legitimacy of considering neurokinin-1 receptor ligands in the design of chimeric drugs is discussed.
Collapse
|
8
|
Abstract
Opioids are the most commonly used and effective analgesic treatments for severe pain, but they have recently come under scrutiny owing to epidemic levels of abuse and overdose. These compounds act on the endogenous opioid system, which comprises four G protein-coupled receptors (mu, delta, kappa, and nociceptin) and four major peptide families (β-endorphin, enkephalins, dynorphins, and nociceptin/orphanin FQ). In this review, we first describe the functional organization and pharmacology of the endogenous opioid system. We then summarize current knowledge on the signaling mechanisms by which opioids regulate neuronal function and neurotransmission. Finally, we discuss the loci of opioid analgesic action along peripheral and central pain pathways, emphasizing the pain-relieving properties of opioids against the affective dimension of the pain experience.
Collapse
Affiliation(s)
- Gregory Corder
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, California 94304, USA; .,Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94304, USA.,Department of Neurosurgery, Stanford University, Palo Alto, California 94304, USA.,Stanford Neurosciences Institute, Palo Alto, California 94304, USA
| | - Daniel C Castro
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63130, USA;
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63130, USA; .,Division of Basic Research, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63130, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, California 94304, USA; .,Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94304, USA.,Department of Neurosurgery, Stanford University, Palo Alto, California 94304, USA.,Stanford Neurosciences Institute, Palo Alto, California 94304, USA.,New York Stem Cell Foundation - Robertson Investigator, Stanford University, Palo Alto, California 94304, USA
| |
Collapse
|
9
|
Wang D, Tawfik VL, Corder G, Low SA, François A, Basbaum AI, Scherrer G. Functional Divergence of Delta and Mu Opioid Receptor Organization in CNS Pain Circuits. Neuron 2018; 98:90-108.e5. [PMID: 29576387 PMCID: PMC5896237 DOI: 10.1016/j.neuron.2018.03.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 01/19/2018] [Accepted: 03/02/2018] [Indexed: 12/21/2022]
Abstract
Cellular interactions between delta and mu opioid receptors (DORs and MORs), including heteromerization, are thought to regulate opioid analgesia. However, the identity of the nociceptive neurons in which such interactions could occur in vivo remains elusive. Here we show that DOR-MOR co-expression is limited to small populations of excitatory interneurons and projection neurons in the spinal cord dorsal horn and unexpectedly predominates in ventral horn motor circuits. Similarly, DOR-MOR co-expression is rare in parabrachial, amygdalar, and cortical brain regions processing nociceptive information. We further demonstrate that in the discrete DOR-MOR co-expressing nociceptive neurons, the two receptors internalize and function independently. Finally, conditional knockout experiments revealed that DORs selectively regulate mechanical pain by controlling the excitability of somatostatin-positive dorsal horn interneurons. Collectively, our results illuminate the functional organization of DORs and MORs in CNS pain circuits and reappraise the importance of DOR-MOR cellular interactions for developing novel opioid analgesics.
Collapse
MESH Headings
- Animals
- Anterior Horn Cells/chemistry
- Anterior Horn Cells/metabolism
- Anterior Horn Cells/pathology
- Central Nervous System/chemistry
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Nerve Net/chemistry
- Nerve Net/metabolism
- Nerve Net/pathology
- Pain/metabolism
- Pain/pathology
- Pain Measurement/methods
- Posterior Horn Cells/chemistry
- Posterior Horn Cells/metabolism
- Posterior Horn Cells/pathology
- Receptors, Opioid, delta/biosynthesis
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Dong Wang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA 94304, USA; Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94304, USA; Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA 94304, USA; Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94304, USA; Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Gregory Corder
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA 94304, USA; Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94304, USA; Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Sarah A Low
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA 94304, USA; Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94304, USA; Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Amaury François
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA 94304, USA; Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94304, USA; Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Allan I Basbaum
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA 94304, USA; Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA 94304, USA; Stanford Neurosciences Institute, Stanford University, Palo Alto, CA 94304, USA; New York Stem Cell Foundation - Robertson Investigator, Stanford University, Palo Alto, CA 94304, USA.
| |
Collapse
|
10
|
MCRT, a chimeric peptide based on morphiceptin and PFRTic-NH2, regulates the depressor effects induced by endokinin A/B. Eur J Pharmacol 2016; 792:33-37. [DOI: 10.1016/j.ejphar.2016.10.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 10/21/2016] [Accepted: 10/21/2016] [Indexed: 01/13/2023]
|
11
|
He C, Gong J, Yang L, Zhang H, Dong S, Zhou L. Pain regulation of endokinin A/B or endokinin C/D on chimeric peptide MCRT in mice. Can J Physiol Pharmacol 2016; 94:955-60. [PMID: 27285300 DOI: 10.1139/cjpp-2015-0554] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study focused on the interactive pain regulation of endokinin A/B (EKA/B, the common C-terminal decapeptide in EKA and EKB) or endokinin C/D (EKC/D, the common C-terminal duodecapeptide in EKC and EKD) on chimeric peptide MCRT (YPFPFRTic-NH2, based on YPFP-NH2 and PFRTic-NH2) at the supraspinal level in mice. Results demonstrated that the co-injection of nanomolar EKA/B and MCRT showed moderate regulation, whereas 30 pmol EKA/B had no effect on MCRT. The combination of EKC/D and MCRT produced enhanced antinociception, which was nearly equal to the sum of the mathematical values of single EKC/D and MCRT. Mechanism studies revealed that pre-injected naloxone attenuated the combination significantly compared with the equivalent analgesic effects of EKC/D alone, suggesting that EKC/D and MCRT might act on two totally independent pathways. Moreover, based on the above results and previous reports, we made two reasonable hypotheses to explain the cocktail-induced analgesia, which may potentially pave the way to explore the respective regulatory mechanisms of EKA/B, EKC/D, and MCRT and to better understand the complicated pain regulation of NK1 and μ opioid receptors, as follows: (1) MCRT and endomorphin-1 possibly activated different μ subtypes; and (2) picomolar EKA/B might motivate the endogenous NPFF system after NK1 activation.
Collapse
Affiliation(s)
- Chunbo He
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, 222 Tianshui South Road, Lanzhou 730000, China
| | - Junbin Gong
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, 222 Tianshui South Road, Lanzhou 730000, China
| | - Lixia Yang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, 222 Tianshui South Road, Lanzhou 730000, China
| | - Hongwei Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, 222 Tianshui South Road, Lanzhou 730000, China
| | - Shouliang Dong
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, 222 Tianshui South Road, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 222 Tianshui South Road, Lanzhou 730000, China
| | - Lanxia Zhou
- The Core Laboratory of the First Affiliated Hospital, Lanzhou University, 1 Donggang West Road, Lanzhou 730000, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, Lanzhou 730000, China
| |
Collapse
|
12
|
Biological evaluation and molecular docking studies of AA3052, a compound containing a μ-selective opioid peptide agonist DALDA and d-Phe-Phe-d-Phe-Leu-Leu-NH2, a substance P analogue. Eur J Pharm Sci 2016; 93:11-20. [PMID: 27423260 DOI: 10.1016/j.ejps.2016.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/05/2016] [Accepted: 07/10/2016] [Indexed: 11/20/2022]
Abstract
The design of novel drugs for pain relief with improved analgesic properties and diminished side effect induction profile still remains a challenging pursuit. Tolerance is one of the most burdensome phenomena that may hamper ongoing opioid therapy, especially in chronic pain patients. Therefore, a promising strategy of hybridizing two pharmacophores that target distinct binding sites involved in pain modulation and transmission was established. Previous studies have led to the development of opioid agonist/NK1 agonist hybrids that produce sufficient analgesia and also suppress opioid-induced tolerance development. In our present investigation we assessed the antinociceptive potency of a new AA3052 chimera comprised of a potent MOR selective dermorphin derivative (DALDA) and an NK1 agonist, a stabilized substance P analogue. We have shown that AA3052 significantly prolonged responses to both mechanical and noxious thermal stimuli in rats after intracerebroventricular administration. Additionally, AA3052 did not trigger the development of tolerance in a 6-day daily injection paradigm nor did it produce any sedative effects, as assessed in the rotarod performance test. However, the antinociceptive effect of AA3052 was independent of opioid receptor stimulation by the DALDA pharmacophore as shown in the agonist-stimulated G-protein assay. Altogether the current results confirm the antinociceptive effectiveness of a novel opioid/SP hybrid agonist, AA3052, and more importantly its ability to inhibit the development of tolerance.
Collapse
|
13
|
Bowman SL, Soohoo AL, Shiwarski DJ, Schulz S, Pradhan AA, Puthenveedu MA. Cell-autonomous regulation of Mu-opioid receptor recycling by substance P. Cell Rep 2015; 10:1925-36. [PMID: 25801029 PMCID: PMC4494997 DOI: 10.1016/j.celrep.2015.02.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 10/31/2014] [Accepted: 02/18/2015] [Indexed: 10/26/2022] Open
Abstract
How neurons coordinate and reprogram multiple neurotransmitter signals is an area of broad interest. Here, we show that substance P (SP), a neuropeptide associated with inflammatory pain, reprograms opioid receptor recycling and signaling. SP, through activation of the neurokinin 1 (NK1R) receptor, increases the post-endocytic recycling of the mu-opioid receptor (MOR) in trigeminal ganglion (TG) neurons in an agonist-selective manner. SP-mediated protein kinase C (PKC) activation is both required and sufficient for increasing recycling of exogenous and endogenous MOR in TG neurons. The target of this cross-regulation is MOR itself, given that mutation of either of two PKC phosphorylation sites on MOR abolishes the SP-induced increase in recycling and resensitization. Furthermore, SP enhances the resensitization of fentanyl-induced, but not morphine-induced, antinociception in mice. Our results define a physiological pathway that cross-regulates opioid receptor recycling via direct modification of MOR and suggest a mode of homeostatic interaction between the pain and analgesic systems.
Collapse
|
14
|
Fujita W, Gomes I, Devi LA. Revolution in GPCR signalling: opioid receptor heteromers as novel therapeutic targets: IUPHAR review 10. Br J Pharmacol 2015; 171:4155-76. [PMID: 24916280 DOI: 10.1111/bph.12798] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/27/2014] [Accepted: 04/16/2014] [Indexed: 12/24/2022] Open
Abstract
GPCRs can interact with each other to form homomers or heteromers. Homomers involve interactions with the same receptor type while heteromers involve interactions between two different GPCRs. These receptor-receptor interactions modulate not only the binding but also the signalling and trafficking properties of individual receptors. Opioid receptor heteromerization has been extensively investigated with the objective of identifying novel therapeutic targets that are as potent as morphine but without the side effects associated with chronic morphine use. In this context, studies have described heteromerization between the different types of opioid receptors and between opioid receptors and a wide range of GPCRs including adrenoceptors, cannabinoid, 5-HT, metabotropic glutamate and sensory neuron-specific receptors. Recent advances in the field involving the generation of heteromer-specific reagents (antibodies or ligands) or of membrane-permeable peptides that disrupt the heteromer interaction are helping to elucidate the physiological role of opioid receptor heteromers and the contribution of the partner receptor to the side effects associated with opioid use. For example, studies using membrane-permeable peptides targeting the heteromer interface have implicated μ and δ receptor heteromers in the development of tolerance to morphine, and heteromers of μ and gastrin-releasing peptide receptors in morphine-induced itch. In addition, a number of ligands that selectively target opioid receptor heteromers exhibit potent antinociception with a decrease in the side effects commonly associated with morphine use. In this review, we summarize the latest findings regarding the biological and functional characteristics of opioid receptor heteromers both in vitro and in vivo.
Collapse
Affiliation(s)
- Wakako Fujita
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | |
Collapse
|
15
|
Taylor BK, Corder G. Endogenous analgesia, dependence, and latent pain sensitization. Curr Top Behav Neurosci 2014; 20:283-325. [PMID: 25227929 PMCID: PMC4464817 DOI: 10.1007/7854_2014_351] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Endogenous activation of µ-opioid receptors (MORs) provides relief from acute pain. Recent studies have established that tissue inflammation produces latent pain sensitization (LS) that is masked by spinal MOR signaling for months, even after complete recovery from injury and re-establishment of normal pain thresholds. Disruption with MOR inverse agonists reinstates pain and precipitates cellular, somatic, and aversive signs of physical withdrawal; this phenomenon requires N-methyl-D-aspartate receptor-mediated activation of calcium-sensitive adenylyl cyclase type 1 (AC1). In this review, we present a new conceptual model of the transition from acute to chronic pain, based on the delicate balance between LS and endogenous analgesia that develops after painful tissue injury. First, injury activates pain pathways. Second, the spinal cord establishes MOR constitutive activity (MORCA) as it attempts to control pain. Third, over time, the body becomes dependent on MORCA, which paradoxically sensitizes pain pathways. Stress or injury escalates opposing inhibitory and excitatory influences on nociceptive processing as a pathological consequence of increased endogenous opioid tone. Pain begets MORCA begets pain vulnerability in a vicious cycle. The final result is a silent insidious state characterized by the escalation of two opposing excitatory and inhibitory influences on pain transmission: LS mediated by AC1 (which maintains the accelerator) and pain inhibition mediated by MORCA (which maintains the brake). This raises the prospect that opposing homeostatic interactions between MORCA analgesia and latent NMDAR-AC1-mediated pain sensitization creates a lasting vulnerability to develop chronic pain. Thus, chronic pain syndromes may result from a failure in constitutive signaling of spinal MORs and a loss of endogenous analgesic control. An overarching long-term therapeutic goal of future research is to alleviate chronic pain by either (a) facilitating endogenous opioid analgesia, thus restricting LS within a state of remission, or (b) extinguishing LS altogether.
Collapse
Affiliation(s)
- Bradley K Taylor
- Department of Physiology, School of Medicine, University of Kentucky Medical Center, Lexington, KY, 40536-0298, USA,
| | | |
Collapse
|
16
|
Chabot-Doré AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic synergy between opioid and α2 -adrenoceptors. Br J Pharmacol 2014; 172:388-402. [PMID: 24641506 DOI: 10.1111/bph.12695] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 03/08/2014] [Accepted: 03/12/2014] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Opioid and α2 -adrenoceptor agonists are potent analgesic drugs and their analgesic effects can synergize when co-administered. These supra-additive interactions are potentially beneficial clinically; by increasing efficacy and/or reducing the total drug required to produce sufficient pain relief, undesired side effects can be minimized. However, combination therapies of opioids and α2 -adrenoceptor agonists remain underutilized clinically, in spite of a large body of preclinical evidence describing their synergistic interaction. One possible obstacle to the translation of preclinical findings to clinical applications is a lack of understanding of the mechanisms underlying the synergistic interactions between these two drug classes. In this review, we provide a detailed overview of the interactions between different opioid and α2 -adrenoceptor agonist combinations in preclinical studies. These studies have identified the spinal cord as an important site of action of synergistic interactions, provided insights into which receptors mediate these interactions and explored downstream signalling events enabling synergy. It is now well documented that the activation of both μ and δ opioid receptors can produce synergy with α2 -adrenoceptor agonists and that α2 -adrenoceptor agonists can mediate synergy through either the α2A or the α2C adrenoceptor subtypes. Current hypotheses surrounding the cellular mechanisms mediating opioid-adrenoceptor synergy, including PKC signalling and receptor oligomerization, and the evidence supporting them are presented. Finally, the implications of these findings for clinical applications and drug discovery are discussed. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- A-J Chabot-Doré
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | | | | |
Collapse
|
17
|
Lennon FE, Mirzapoiazova T, Mambetsariev B, Poroyko VA, Salgia R, Moss J, Singleton PA. The Mu opioid receptor promotes opioid and growth factor-induced proliferation, migration and Epithelial Mesenchymal Transition (EMT) in human lung cancer. PLoS One 2014; 9:e91577. [PMID: 24662916 PMCID: PMC3963855 DOI: 10.1371/journal.pone.0091577] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 02/13/2014] [Indexed: 01/08/2023] Open
Abstract
Recent epidemiologic studies implying differences in cancer recurrence based on anesthetic regimens raise the possibility that the mu opioid receptor (MOR) can influence cancer progression. Based on our previous observations that overexpression of MOR in human non-small cell lung cancer (NSCLC) cells increased tumor growth and metastasis, this study examined whether MOR regulates growth factor receptor signaling and epithelial mesenchymal transition (EMT) in human NSCLC cells. We utilized specific siRNA, shRNA, chemical inhibitors and overexpression vectors in human H358 NSCLC cells that were either untreated or treated with various concentrations of DAMGO, morphine, fentanyl, EGF or IGF. Cell function assays, immunoblot and immunoprecipitation assays were then performed. Our results indicate MOR regulates opioid and growth factor-induced EGF receptor signaling (Src, Gab-1, PI3K, Akt and STAT3 activation) which is crucial for consequent human NSCLC cell proliferation and migration. In addition, human NSCLC cells treated with opioids, growth factors or MOR overexpression exhibited an increase in snail, slug and vimentin and decrease ZO-1 and claudin-1 protein levels, results consistent with an EMT phenotype. Further, these effects were reversed with silencing (shRNA) or chemical inhibition of MOR, Src, Gab-1, PI3K, Akt and STAT3 (p<0.05). Our data suggest a possible direct effect of MOR on opioid and growth factor-signaling and consequent proliferation, migration and EMT transition during lung cancer progression. Such an effect provides a plausible explanation for the epidemiologic findings.
Collapse
Affiliation(s)
- Frances E. Lennon
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Tamara Mirzapoiazova
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Bolot Mambetsariev
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Valeriy A. Poroyko
- Department of Surgery, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Ravi Salgia
- Section of Hematology/Oncology, Department of Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Jonathan Moss
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Patrick A. Singleton
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
18
|
Gomes I, Fujita W, Chandrakala MV, Devi LA. Disease-specific heteromerization of G-protein-coupled receptors that target drugs of abuse. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 117:207-65. [PMID: 23663971 DOI: 10.1016/b978-0-12-386931-9.00009-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Drugs of abuse such as morphine or marijuana exert their effects through the activation of G-protein-coupled receptors (GPCRs), the opioid and cannabinoid receptors, respectively. Moreover, interactions between either of these receptors have been shown to be involved in the rewarding effects of drugs of abuse. Recent advances in the field, using a variety of approaches, have demonstrated that many GPCRs, including opioid, cannabinoid, and dopamine receptors, can form associations between different receptor subtypes or with other GPCRs to form heteromeric complexes. The formation of these complexes, in turn, leads to the modulation of the properties of individual protomers. The development of tools that can selectively disrupt GPCR heteromers as well as monoclonal antibodies that can selectively block signaling by specific heteromer pairs has indicated that heteromers involving opioid, cannabinoid, or dopamine receptors may play a role in various disease states. In this review, we describe evidence for opioid, cannabinoid, and dopamine receptor heteromerization and the potential role of GPCR heteromers in pathophysiological conditions.
Collapse
Affiliation(s)
- Ivone Gomes
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, USA
| | | | | | | |
Collapse
|
19
|
Lee CWS, Ho IK. Pharmacological Profiles of Oligomerized μ-Opioid Receptors. Cells 2013; 2:689-714. [PMID: 24709876 PMCID: PMC3972655 DOI: 10.3390/cells2040689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/30/2013] [Accepted: 10/09/2013] [Indexed: 01/21/2023] Open
Abstract
Opioids are widely prescribed pain relievers with multiple side effects and potential complications. They produce analgesia via G-protein-protein coupled receptors: μ-, δ-, κ-opioid and opioid receptor-like 1 receptors. Bivalent ligands targeted to the oligomerized opioid receptors might be the key to developing analgesics without undesired side effects and obtaining effective treatment for opioid addicts. In this review we will update the biological effects of μ-opioids on homo- or hetero-oligomerized μ-opioid receptor and discuss potential mechanisms through which bivalent ligands exert beneficial effects, including adenylate cyclase regulation and receptor-mediated signaling pathways.
Collapse
Affiliation(s)
- Cynthia Wei-Sheng Lee
- Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Ing-Kang Ho
- Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung 40447, Taiwan.
| |
Collapse
|
20
|
Largent-Milnes TM, Brookshire SW, Skinner DP, Hanlon KE, Giuvelis D, Yamamoto T, Davis P, Campos CR, Nair P, Deekonda S, Bilsky EJ, Porreca F, Hruby VJ, Vanderah TW. Building a better analgesic: multifunctional compounds that address injury-induced pathology to enhance analgesic efficacy while eliminating unwanted side effects. J Pharmacol Exp Ther 2013; 347:7-19. [PMID: 23860305 PMCID: PMC3781412 DOI: 10.1124/jpet.113.205245] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 07/08/2013] [Indexed: 12/27/2022] Open
Abstract
The most highly abused prescription drugs are opioids used for the treatment of pain. Physician-reported drug-seeking behavior has resulted in a significant health concern among doctors trying to adequately treat pain while limiting the misuse or diversion of pain medications. In addition to abuse liability, opioid use is associated with unwanted side effects that complicate pain management, including opioid-induced emesis and constipation. This has resulted in restricting long-term doses of opioids and inadequate treatment of both acute and chronic debilitating pain, demonstrating a compelling need for novel analgesics. Recent reports indicate that adaptations in endogenous substance P/neurokinin-1 receptor (NK1) are induced by chronic pain and sustained opioid exposure, and these changes may contribute to processes responsible for opioid abuse liability, emesis, and analgesic tolerance. Here, we describe a multifunctional mu-/delta-opioid agonist/NK1 antagonist compound [Tyr-d-Ala-Gly-Phe-Met-Pro-Leu-Trp-NH-Bn(CF3)2 (TY027)] that has a preclinical profile of excellent antinociceptive efficacy, low abuse liability, and no opioid-related emesis or constipation. In rodent models of acute and neuropathic pain, TY027 demonstrates analgesic efficacy following central or systemic administration with a plasma half-life of more than 4 hours and central nervous system penetration. These data demonstrate that an innovative opioid designed to contest the pathology created by chronic pain and sustained opioids results in antinociceptive efficacy in rodent models, with significantly fewer side effects than morphine. Such rationally designed, multitargeted compounds are a promising therapeutic approach in treating patients who suffer from acute and chronic pain.
Collapse
Affiliation(s)
- T M Largent-Milnes
- Department of Pharmacology (T.M.L.-M., S.W.B., D.P.S., K.E.H., P.D., C.R.C., F.P., T.W.V.), and Department of Chemistry (T.Y., P.N, S.D., V.J.H.), University of Arizona, Tucson, Arizona; and Center for Excellence in Neuroscience, University of New England, Biddeford, Maine (K.E.H., D.G., E.J.B., F.P., T.W.V.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wang G, Coleman CG, Chan J, Faraco G, Marques-Lopes J, Milner TA, Guruju MR, Anrather J, Davisson RL, Iadecola C, Pickel VM. Angiotensin II slow-pressor hypertension enhances NMDA currents and NOX2-dependent superoxide production in hypothalamic paraventricular neurons. Am J Physiol Regul Integr Comp Physiol 2013; 304:R1096-106. [PMID: 23576605 DOI: 10.1152/ajpregu.00367.2012] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adaptive changes in glutamatergic signaling within the hypothalamic paraventricular nucleus (PVN) may play a role in the neurohumoral dysfunction underlying the hypertension induced by "slow-pressor" ANG II infusion. We hypothesized that these adaptive changes alter production of gp91phox NADPH oxidase (NOX)-derived reactive oxygen species (ROS) or nitric oxide (NO), resulting in enhanced glutamatergic signaling in the PVN. Electron microscopic immunolabeling showed colocalization of NOX2 and N-methyl-D-aspartate receptor (NMDAR) NR1 subunits in PVN dendrites, an effect enhanced (+48%, P < 0.05 vs. saline) in mice receiving ANG II (600 ng·kg⁻¹·min⁻¹ sc). Isolated PVN cells or spinally projecting PVN neurons from ANG II-infused mice had increased levels of ROS at baseline (+40 ± 5% and +57.6 ± 7.7%, P < 0.01 vs. saline) and after NMDA (+24 ± 7% and +17 ± 5.5%, P < 0.01 and P < 0.05 vs. saline). In contrast, ANG II infusion suppressed NO production in PVN cells at baseline (-29.1 ± 5.2%, P < 0.05 vs. saline) and after NMDA (-18.9 ± 2%, P < 0.01 vs. saline), an effect counteracted by NOX inhibition. In whole cell recording of unlabeled and spinally labeled PVN neurons in slices, NMDA induced a larger inward current in ANG II than in saline groups (+79 ± 24% and +82.9 ± 6.6%, P < 0.01 vs. saline), which was reversed by the ROS scavenger MnTBAP and the NO donor S-nitroso-N-acetylpenicillamine (P > 0.05 vs. control). These findings suggest that slow-pressor ANG II increases the association of NR1 with NOX2 in dendrites of PVN neurons, resulting in enhanced NOX-derived ROS and reduced NO during glutamatergic activity. The resulting enhancement of NMDAR activity may contribute to the neurohumoral dysfunction underlying the development of slow-pressor ANG II hypertension.
Collapse
Affiliation(s)
- Gang Wang
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
López-Bellido R, Barreto-Valer K, Rodríguez RE. Expression of tachykinin receptors (tacr1a and tacr1b) in zebrafish: influence of cocaine and opioid receptors. J Mol Endocrinol 2013; 50:115-29. [PMID: 23256992 DOI: 10.1530/jme-12-0199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Opioid and tachykinin receptors (TACRs) are closely related in addiction and pain processes. In zebrafish, opioid receptors have been cloned and characterized both biochemically and pharmacologically. However, the tacr1 gene has not yet been described in zebrafish. The aim of this research was to identify the tacr1 gene, study the effects of cocaine on tacr1, and analyze the interaction between tacr1 and opioid receptors. We have identified a duplicate of tacr1 gene in zebrafish, designated as tacr1a and tacr1b. Phylogenetic analyses revealed an alignment of these receptors in the Tacr1 fish cluster, with a clear distinction from other TACR1s of amphibians, birds, and mammals. Our qPCR results showed that tacr1a and tacr1b mRNAs are expressed during embryonic development. Whole-mount in situ hybridization showed tacr1 expression in the CNS and in the peripheral tissues. Cocaine (1.5 μM) induced an upregulation of tacr1a and tacr1b at 24 and 48 h post-fertilization (hpf; except for tacr1a at 48 hpf, which was downregulated). By contrast, HEK-293 cells transfected with tacr1a and tacr1b and exposed to cocaine showed a downregulation of tacr1s. The knockdown of ZfDOR2 and ZfMOR, opioid receptors, induced a down- and upregulation of tacr1a and tacr1b respectively. In conclusion, tacr1a and tacr1b in zebrafish are widely expressed throughout the CNS and peripherally, suggesting a critical role of these tacr1s during embryogenesis. tacr1a and tacr1b mRNA expression is altered by cocaine exposure and by the knockdown of opioid receptors. Thus, zebrafish can provide clues for a better understanding of the relationship between tachykinin and opioid receptors in pain and addiction during embryonic development.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line
- Cloning, Molecular
- Cocaine/pharmacology
- Gene Expression Regulation
- Gene Expression Regulation, Developmental/drug effects
- Gene Knockdown Techniques
- HEK293 Cells
- Humans
- Molecular Sequence Data
- Phylogeny
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Drug/genetics
- Receptors, Drug/metabolism
- Receptors, Opioid/genetics
- Receptors, Opioid/metabolism
- Receptors, Tachykinin/classification
- Receptors, Tachykinin/genetics
- Sequence Alignment
- Transfection
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
Collapse
Affiliation(s)
- Roger López-Bellido
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
| | | | | |
Collapse
|
23
|
Walsh SL, Heilig M, Nuzzo PA, Henderson P, Lofwall MR. Effects of the NK1 antagonist, aprepitant, on response to oral and intranasal oxycodone in prescription opioid abusers. Addict Biol 2013; 18:332-43. [PMID: 22260216 DOI: 10.1111/j.1369-1600.2011.00419.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Pre-clinical studies suggest that the neurokinin-1 (NK1) receptor may modulate the response to opioids, with NK(1) inactivation leading to decreased opioid reinforcement, tolerance and withdrawal. Aprepitant is a selective NK1 antagonist currently marketed for clinical use as an anti-emetic. This 6-week in-patient study used a randomized, double-blind, double-dummy, within-subject, crossover design. Subjects (n = 8; 6 male/2 female) were healthy, adult volunteers who provided subjective and objective evidence of current prescription opioid abuse (without physical dependence) and underwent careful medical and psychiatric screening. Fifteen experimental conditions, consisting of one aprepitant dose (0, 40 and 200 mg, p.o. given as a 2-hour pre-treatment) in combination with one oxycodone dose [placebo, oral (20 and 40 mg/70 kg) and intranasal (15 and 30 mg/70 kg)], were examined. Sessions were conducted at least 48-hour apart and multi-dimensional measures were collected repeatedly throughout the 6-hour session duration. Oxycodone, by both routes of administration, produced significant dose-related effects on the predicted measures (e.g. subjective measures of abuse liability, respiratory depression and miosis). Pre-treatment with aprepitant (200 mg) significantly enhanced ratings of oxycodone subjective effects related to euphoria and liking and doubled the street value estimates for the highest test doses of oxycodone by both routes. Some objective measures (respiratory function, observer-rated opioid agonist effects) were similarly enhanced by pre-treatment with the highest dose of aprepitant. All dose combinations were safely tolerated. These findings are discussed in the context of the potential utility of NK1 antagonists in the treatment of opioid use disorders.
Collapse
Affiliation(s)
- Sharon L Walsh
- Center on Drug and Alcohol Research, University of Kentucky, Lexington, KY 40503, USA.
| | | | | | | | | |
Collapse
|
24
|
Wilson-Poe AR, Morgan MM, Aicher SA, Hegarty DM. Distribution of CB1 cannabinoid receptors and their relationship with mu-opioid receptors in the rat periaqueductal gray. Neuroscience 2012; 213:191-200. [PMID: 22521830 DOI: 10.1016/j.neuroscience.2012.03.038] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 02/15/2012] [Accepted: 03/13/2012] [Indexed: 11/15/2022]
Abstract
The periaqueductal gray (PAG) is part of a descending pain modulatory system that, when activated, produces widespread and profound antinociception. Microinjection of either opioids or cannabinoids into the PAG elicits antinociception. Moreover, microinjection of the cannabinoid 1 (CB1) receptor agonist HU-210 into the PAG enhances the antinociceptive effect of subsequent morphine injections, indicating a direct relationship between these two systems. The objective of this study was to characterize the distribution of CB1 receptors in the dorsolateral and ventrolateral PAG in relationship to mu-opioid peptide (MOP) receptors. Immunocytochemical analysis revealed extensive and diffuse CB1 receptor labeling in the PAG, 60% of which was found in somatodendritic profiles. CB1 and MOP receptor immunolabeling were co-localized in 32% of fluorescent Nissl-stained cells that were analyzed. Eight percent (8%) of PAG neurons that were MOP receptor-immunoreactive (-ir) received CB1 receptor-ir appositions. Ultrastructural analysis confirmed the presence of CB1 receptor-ir somata, dendrites and axon terminals in the PAG. These results indicate that behavioral interactions between cannabinoids and opioids may be the result of cellular adaptations within PAG neurons co-expressing CB1 and MOP receptors.
Collapse
Affiliation(s)
- A R Wilson-Poe
- Department of Neuroscience, Washington State University, 14204 NE Salmon Creek Avenue, Vancouver, WA 98686, USA.
| | | | | | | |
Collapse
|
25
|
Parenti C, Aricò G, Ronsisvalle G, Scoto GM. Supraspinal injection of Substance P attenuates allodynia and hyperalgesia in a rat model of inflammatory pain. Peptides 2012; 34:412-8. [PMID: 22306475 DOI: 10.1016/j.peptides.2012.01.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 01/20/2012] [Accepted: 01/20/2012] [Indexed: 01/03/2023]
Abstract
The neuropeptide Substance P (SP), that has a high affinity for the neurokinin 1 (NK1) receptor, is involved in modulation of pain transmission. Although SP is thought to have excitatory actions and promote nociception in the spinal cord, the peptide induces analgesia at the supraspinal level. The aim of this study was to evaluate the role of supraspinal SP and the NK1 receptor in inflammatory pain induced by injection of carrageenan in the hind paw of the rat. There are two nociceptive behavioral responses associated with this pain state: mechanical allodynia and heat hyperalgesia. Because the NK1 receptor colocalizes with the MOP receptor in supraspinal sites involved in pain modulation, we also decided to study the possible involvement of the opioid system on SP-induced analgesia. We found that treatment with SP, at doses of 3.5, 5 and 7 μg/5 μl/rat i.c.v., clearly showed inhibition of allodynia and hyperalgesia. Pretreatment with the selective NK1 antagonist L-733,060 (10mg/kg i.p.) blocked the SP-induced analgesia, suggesting the involvement of the NK1 receptor. This SP-induced analgesia was significantly reduced by administration of the opioid antagonist naloxone (3mg/kg s.c.). This reduction occurred when SP was administered either before or after the carrageenan injection. These results suggest a significant antinociceptive role for SP and the NK1 receptor in inflammatory pain at the supraspinal level, possibly through the release of endogenous opioids.
Collapse
Affiliation(s)
- Carmela Parenti
- Department of Drug Sciences-Pharmacology and Toxicology Section, University of Catania, v.le A. Doria 6, 95125 Catania, Italy.
| | | | | | | |
Collapse
|
26
|
Sandkühler J, Ruscheweyh R. Opioids and central sensitisation: I. Pre-emptive analgesia. Eur J Pain 2012; 9:145-8. [PMID: 15737804 DOI: 10.1016/j.ejpain.2004.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Accepted: 05/17/2004] [Indexed: 11/20/2022]
Abstract
Opioids are powerful analgesics when used to treat acute pain and some forms of chronic pain. A large body of literature has shown that opioids can, in addition, also prevent (this review) or induce and perhaps reverse, some forms central sensitisation in in vitro and in vivo animal models of pain. However, the concept of central sensitisation is, at present, ambiguous and the usefulness of opioids as preemptive analgesics in human pain patients is still not clear.
Collapse
Affiliation(s)
- Jürgen Sandkühler
- Department of Neurophysiology, Centre for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria.
| | | |
Collapse
|
27
|
Jin Q, Lu L, Yang Y, Dong S. Effects of endokinin A/B, endokinin C/D, and endomorphin-1 on the regulation of mean arterial blood pressure in rats. Peptides 2011; 32:2428-35. [PMID: 22037058 DOI: 10.1016/j.peptides.2011.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/16/2011] [Accepted: 10/16/2011] [Indexed: 11/18/2022]
Abstract
Endokinins are four novel human tachykinins, including endokinins A (EKA), B (EKB), C (EKC), and D (EKD). Endokinin A/B (EKA/B) is the common C-terminal decapeptide in EKA and EKB, while endokinin C/D (EKC/D) is the common C-terminal duodecapeptide in EKC and EKD. In this study, we attempted to investigate the interactions between EKA/B, EKC/D, and endomorphin-1 (EM-1) on the depressor effect at peripheral level. The effects of EKA/B produced a U-shaped curve. The maximal effect was caused by 10 nmol/kg. EKC/D and EM-1 showed a dose-dependent relationship. Co-administration of EKA/B (0.1, 1, 10 nmol/kg) with EM-1 produced effects similar to those of EKA/B alone but slightly lower. Co-injection of EKA/B (100 nmol/kg) with EM-1 caused an effect stronger than any separate injection. Co-administration of EKC/D (10 nmol/kg) with EM-1 (30 nmol/kg) caused a depressor effect, which was one of the tradeoffs of EM-1 and EKC/D. Mechanism studies showed that SR140333B could block the depressor effects of EKA/B, EKC/D, EM-1, EKA/B+EM-1, and EKC/D+EM-1; SR48968C could block EM-1, EKA/B, EKC/D, and EKC/D+EM-1 and partially block EKA/B+EM-1; SR142801 could block EM-1, EKC/D, and EKC/D+EM-1 and partially block EKA/B and EKA/B+EM-1; naloxone could block EM-1, EKC/D, and EKC/D+EM-1 and partially block EKA/B and EKA/B+EM-1. Pretreatment with NG-nitro-l-arginine methyl ester partially decreased depressor intensity and half-recovery time of EKA/B and EKC/D.
Collapse
Affiliation(s)
- Qiaoying Jin
- Institute of Biochemistry and Molecular Biology, School of Life Sciences
| | | | | | | |
Collapse
|
28
|
Yang Y, Ni Z, Dong S. Effects of Endokinin A/B and Endokinin C/D on the antinociception of Endomorphin-1 in mice. Peptides 2010; 31:689-95. [PMID: 20035812 DOI: 10.1016/j.peptides.2009.12.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 12/16/2009] [Accepted: 12/16/2009] [Indexed: 01/24/2023]
Abstract
In our previous study, Endokinin A/B (EKA/B, the common C-terminal decapeptide in Endokinin A and Endokinin B) was found to induce analgesic effect at high dose and nociception at low dose, while Endokinin C/D (EKC/D, the common C-terminal duodecapeptide in Endokinin C and Endokinin D) has analgesic effect only. So in this study an attempt was undertaken to investigate the interaction of EKA/B and EKC/D with Endomorphin-1 (EM-1) on antinociceptive effect at supraspinal level. Results showed that the antinociceptive effect of EM-1 was enhanced by high dose of EKA/B and abolished by low dose of EKA/B, while EKC/D could only enhance the analgesic effect. Mechanism studies showed that EKA/B blocked the antinociception of EM-1 by activating neurokinin-1 receptor (NK(1)), whose specific antagonist, SR140333B could fully block EKA/B-induced attenuation on the analgesic response of EM-1. Surprisingly, EKC/D could also block the same EKA/B-induced attenuation. Taken together, the different effects of EKA/B and EKC/D on the antinociception of EM-1 may pave the way for a new strategy on investigating the interaction between tachykinins and opioids on pain modulation.
Collapse
Affiliation(s)
- Yinliang Yang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
| | | | | |
Collapse
|
29
|
Yang Y, Dong S. Effects of Endokinin A/B and Endokinin C/D on the modulation of pain in mice. Peptides 2010; 31:94-100. [PMID: 19854230 DOI: 10.1016/j.peptides.2009.10.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 10/14/2009] [Accepted: 10/14/2009] [Indexed: 10/20/2022]
Abstract
Endokinins are novel tachykinins encoded on the human TAC4 and consist of Endokinin A (EKA), B (EKB), C (EKC) and D (EKD). To date, the function of Endokinins in pain processing was not fully understood. Therefore the aim of this study was to investigate the effects of Endokinin A/B (EKA/B, the common C-terminal decapeptide in EKA and EKB) and Endokinin C/D (EKC/D, the common C-terminal duodecapeptide in EKC and EKD) on pain modulation at supraspinal level in mice. Intracerebroventricular (i.c.v.) administration of EKA/B (1, 3, 12, 20nmol/mouse) dose dependently induced potent analgesic effect. This effect could be fully antagonized by SR140333B but not SR48968C or SR142801. Naloxone could also block the analgesic effect, suggesting that this analgesic effect is related to opioid receptors. However, i.c.v. administration of EKA/B (10, 30, 100pmol/mouse) caused hyperalgesic effect significantly, with a "U" shape curve. Interestingly, the hyperalgesic effect induced by EKA/B could be attenuated by SR140333B, SR142801 but not SR48968C. I.c.v. administration of EKC/D (1, 3, 12, 20nmol/mouse) also dose dependently induced analgesic effect, which could not be blocked by SR48968C or SR142801 or naloxone. But to our astonishment, it could be significantly enhanced by SR140333B. More interestingly, the hyperalgesic effect induced by EKA/B could be significantly attenuated by EKC/D. In addition, the analgesic effect induced by co-administration of EKA/B and EKC/D was much less stronger than the effect of either EKA/B or EKC/D.
Collapse
Affiliation(s)
- Yinliang Yang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | | |
Collapse
|
30
|
Dissociation of the opioid receptor mechanisms that control mechanical and heat pain. Cell 2009; 137:1148-59. [PMID: 19524516 DOI: 10.1016/j.cell.2009.04.019] [Citation(s) in RCA: 365] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Revised: 01/30/2009] [Accepted: 04/01/2009] [Indexed: 12/22/2022]
Abstract
Delta and mu opioid receptors (DORs and MORs) are inhibitory G protein-coupled receptors that reportedly cooperatively regulate the transmission of pain messages by substance P and TRPV1-expressing pain fibers. Using a DOReGFP reporter mouse we now show that the DOR and MOR are, in fact, expressed by different subsets of primary afferents. The MOR is expressed in peptidergic pain fibers, the DOR in myelinated and nonpeptidergic afferents. Contrary to the prevailing view, we demonstrate that the DOR is trafficked to the cell surface under resting conditions, independently of substance P, and internalized following activation by DOR agonists. Finally, we show that the segregated DOR and MOR distribution is paralleled by a remarkably selective functional contribution of the two receptors to the control of mechanical and heat pain, respectively. These results demonstrate that behaviorally relevant pain modalities can be selectively regulated through the targeting of distinct subsets of primary afferent pain fibers.
Collapse
|
31
|
Lin LH, Taktakishvili OM, Talman WT. Colocalization of neurokinin-1, N-methyl-D-aspartate, and AMPA receptors on neurons of the rat nucleus tractus solitarii. Neuroscience 2008; 154:690-700. [PMID: 18479828 DOI: 10.1016/j.neuroscience.2008.03.078] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Revised: 03/28/2008] [Accepted: 03/30/2008] [Indexed: 02/01/2023]
Abstract
Substance P (SP) and glutamate are implicated in cardiovascular regulation by the nucleus tractus solitarii (NTS). Our earlier studies suggest that SP, which acts at neurokinin 1 (NK1) receptors, is not a baroreflex transmitter while glutamate is. On the other hand, our recent studies showed that loss of NTS neurons expressing NK1 receptors leads to loss of baroreflex responses and increased blood pressure lability. Furthermore, studies have suggested that SP may interact with glutamate in the NTS. In this study, we sought to test the hypothesis that NK1 receptors colocalize with glutamate receptors, either N-methyl-d-aspartate (NMDA) receptors or AMPA receptors or both in the NTS. We performed double-label immunofluorescent staining for NK1 receptors and either N-methyl-d-aspartate receptor subunit 1 (NMDAR1) or AMPA specific glutamate receptor subunit 2 (GluR2) in the rat NTS. Because vesicular glutamate transporter 2 (VGLUT2) containing fibers are prominent in portions of the NTS where cardiovascular afferent fibers terminate, we also performed double-label immunofluorescent staining for NK1 receptors and VGLUT2. Confocal microscopic images showed that NK1 receptors-immunoreactivity (IR) and NMDAR1-IR colocalized in the same neurons in many NTS subnuclei. Almost all NTS neurons positive for NK1 receptor-IR also contained NMDAR1-IR, but only 53.4% to 74.8% of NMDAR1-IR positive neurons contained NK1 receptors-IR. NK1 receptor-IR and GluR2-IR also colocalized in many neurons in NTS subnuclei. A majority of NK1 receptor-IR positive NTS neurons also contained GluR2-IR, but only 45.8% to 73.9% of GluR2-IR positive NTS neurons contained NK1 receptors-IR. Our results also showed that fibers labeled for VGLUT2-IR were in close apposition to fibers and neurons labeled for NK1 receptor-IR. The data support our hypothesis, provide an anatomical framework for glutamate and SP interactions, and may explain the loss of baroreflexes when NTS neurons, which could respond to glutamate as well as SP, are killed.
Collapse
MESH Headings
- Animals
- Fluorescent Antibody Technique, Indirect
- Male
- Microscopy, Confocal
- Neurons/metabolism
- Neurons/ultrastructure
- Rats
- Rats, Sprague-Dawley
- Receptors, AMPA/biosynthesis
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, N-Methyl-D-Aspartate/biosynthesis
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, Neurokinin-1/metabolism
- Solitary Nucleus/cytology
- Solitary Nucleus/metabolism
- Solitary Nucleus/ultrastructure
- Vesicular Glutamate Transport Protein 2/biosynthesis
- Vesicular Glutamate Transport Protein 2/genetics
Collapse
Affiliation(s)
- L H Lin
- Department of Neurology, University of Iowa, 1191 Medical Laboratories, Iowa City, IA 52242, USA.
| | | | | |
Collapse
|
32
|
Fu CY, Tang XL, Yang Q, Chen Q, Wang R. Effects of rat/mouse hemokinin-1, a mammalian tachykinin peptide, on the antinociceptive activity of pethidine administered at the peripheral and supraspinal level. Behav Brain Res 2007; 184:39-46. [PMID: 17675256 DOI: 10.1016/j.bbr.2007.06.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 05/14/2007] [Accepted: 06/26/2007] [Indexed: 11/30/2022]
Abstract
We have recently reported that rat/mouse hemokinin-1 (r/m HK-1), a mammalian tachykinin, produced dose- and time-related antinociceptive effects at the supraspinal level via activating NK(1) receptors. Moreover, r/m HK-1 remarkably enhanced both the antinociceptive extent and duration of morphine administered at the peripheral and supraspinal level through a convergence of pharmacological effects of opioid-responsive neurons. Pethidine hydrochloride is an important narcotic analgesic, which acts as an opiate agonist and has pharmacological effects similar to morphine. To improve our knowledge of the pharmacology of pethidine, the aim of the present study was to investigate the relationship between the nociception of r/m HK and pethidine by comparing it with that of r/m HK-1 and morphine. Our data showed that r/m HK-1 remarkably enhanced the antinociceptive extent of pethidine administered at the peripheral level, but not at the supraspinal level. These antinociceptive effects were blocked by prior treatment with the classical opioid receptor antagonist naloxone, indicating that the potentiated analgesic effect is mediated by opioid-responsive neurons. Differences in the antinociceptive activity of pethidine and morphine in combination with r/m HK-1, arise because there are differences in the physicochemical and pharmacokinetic properties of pethidine and morphine, particularly their lipophilicity. Our results may pave the way for a new strategy for the control of pain and may provide a clinical strategy to enable selection of either opioid as a priority.
Collapse
Affiliation(s)
- Cai-Yun Fu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, Lanzhou University, 222 Tian Shui South Road, Lanzhou 730000, People's Republic of China
| | | | | | | | | |
Collapse
|
33
|
Ro JY, Lee J, Capra NF, Zhang Y. Role of soluble guanylate cyclase in the trigeminal subnucleus caudalis in capsaicin-induced muscle hypersensitivity. Brain Res 2007; 1184:141-8. [PMID: 17980861 DOI: 10.1016/j.brainres.2007.09.085] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 09/25/2007] [Accepted: 09/26/2007] [Indexed: 01/07/2023]
Abstract
Nitric oxide (NO) produces its effects by activating soluble guanylate cyclase (sGC). In the present study, we investigated the potential role of sGC in the subnucleus caudalis (Vc) in mediating masseter hypersensitivity under acute inflammatory condition in male Sprague-Dawley rats. First, our Western blot analysis revealed that sGC protein is reliably detected in the Vc. Subsequent immunohistochemical studies demonstrated that neuronal cell bodies in the superficial laminae of the Vc positively stained for sGC. Astrocytes in deeper lamina of the Vc also showed sGC immunoreactivity. We then tested whether intrathecal administration of sGC inhibitors, methylene blue (MB), and ODQ, in the Vc, attenuates masseter hypersensitivity induced by intramuscular injection of capsaicin. Intrathecal MB or ODQ significantly blocked the capsaicin-induced reduction of mechanical threshold to noxious stimulation of the masseter. These data indicate that the NO-sGC pathway in the Vc is involved in mediating orofacial muscle hypersensitivity under acute inflammatory conditions.
Collapse
Affiliation(s)
- Jin Y Ro
- Department of Biomedical Sciences, Program in Neuroscience, University of Maryland Baltimore, School of Dentistry, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
34
|
Kainate receptors are primarily postsynaptic to SP-containing axon terminals in the trigeminal dorsal horn. Brain Res 2007; 1184:149-59. [PMID: 17964552 DOI: 10.1016/j.brainres.2007.09.070] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 09/25/2007] [Accepted: 09/26/2007] [Indexed: 02/05/2023]
Abstract
Kainate receptors (KARs) are involved in the modulation and transmission of nociceptive information from peripheral afferents to neurons in the spinal cord and trigeminal dorsal horns. KARs are found at both pre- and postsynaptic sites in the dorsal horn. We hypothesized that KARs and Substance P (SP), a modulatory neuropeptide that is used as a marker of nociceptive afferents, have a complex interactive relationship. To determine the cellular relationship and connectivity between KARs and SP afferents, we used electron microscopic dual immunocytochemical analysis to examine the ultrastructural localization of KAR subunits GluR5, 6 and 7 (GluR5,6,7) in relation to SP within laminae I and II in the rat trigeminal dorsal horn. KARs were distributed both postsynaptically in dendrites and somata (51% of GluR5,6,7 immunoreactive (-ir) profiles) and presynaptically in axons and axon terminals (45%). We also found GluR5,6,7-ir glial profiles (5%). The majority of SP-ir profiles were presynaptic axons and axon terminals. SP-ir dendritic profiles were rare, yet 23% contained GluR5,6,7 immunoreactivity. GluR5,6,7 and SP were also colocalized at presynaptic sites (18% of GluR5,6,7-ir axons and axon terminals contained SP; while 11% of SP-ir axons and axon terminals contained GluR5,6,7). The most common interaction between KARs and SP we observed was GluR5,6,7-ir dendrites contacted by SP-ir axon terminals; 54% of the dendritic targets of SP-ir axon terminals were GluR5,6,7-ir. These results provide anatomical evidence that KARs primarily mediate nociceptive transmission postsynaptic to SP-containing afferents and may also modulate the presynaptic release of SP and glutamate in trigeminal dorsal horn.
Collapse
|
35
|
Wan Q, Douglas SD, Wang X, Kolson DL, O'Donnell LA, Ho WZ. Morphine upregulates functional expression of neurokinin-1 receptor in neurons. J Neurosci Res 2007; 84:1588-96. [PMID: 16983662 DOI: 10.1002/jnr.21053] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neuronkinin-1 receptor (NK-1R), the neuropeptide substance P (SP) preferring receptor, is highly expressed in areas of the central nervous system (CNS) that are especially implicated in depression, anxiety, and stress. Repeated exposure to opioids may sensitize neuronal systems involved in stress response. We examined the effects of morphine, the principal metabolite of heroin, on the functional expression of NK-1R in the cortical neurons. NK-1R and mu-opioid receptor (MOR) are co-expressed in the cortical neurons. Morphine enhanced NK-1R expression in the cortical neurons at both the mRNA and protein levels. The upregulated NK-1R by morphine had functional activity, because morphine-treated cortical neurons had greater SP-induced Ca(2+) mobilization than untreated neurons. Blocking opioid receptors on the cortical neurons by naltrexone or CTAP (a mu-opioid receptor antagonist) abolished the morphine action. Investigation of the mechanism(s) responsible for the morphine action showed that morphine activated NK-1R promoter and induced the phosphorylation of p38 MAPK protein in the cortical neurons. These in vitro data provide a plausible cellular mechanism for opioid-mediated neurological disorders.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Blotting, Western/methods
- Calcium/metabolism
- Cerebral Cortex/cytology
- Drug Interactions
- Embryo, Mammalian
- Enzyme Activation/drug effects
- Enzyme Inhibitors/pharmacology
- Fluorescent Antibody Technique/methods
- Morphine/pharmacology
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Narcotics/pharmacology
- Neurons/drug effects
- Peptide Fragments
- Peptides/pharmacology
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Receptors, Neurokinin-1/genetics
- Receptors, Neurokinin-1/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Somatostatin
- Up-Regulation/drug effects
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Qi Wan
- Division of Allergy and Immunology, Joseph Stokes Jr. Research Institute at The Children's Hospital of Philadelphia, Department of Pediatrics, Philadelphia, Pennsylvania
| | | | | | | | | | | |
Collapse
|
36
|
Mizoguchi H, Nakayama D, Watanabe H, Ito K, Sakurada W, Sawai T, Fujimura T, Sato T, Sakurada T, Sakurada S. Involvement of spinal μ1-opioid receptors on the Tyr-d-Arg-Phe-sarcosine-induced antinociception. Eur J Pharmacol 2006; 540:67-72. [PMID: 16730704 DOI: 10.1016/j.ejphar.2006.04.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 04/22/2006] [Accepted: 04/25/2006] [Indexed: 12/27/2022]
Abstract
The involvement of spinal mu-opioid receptor subtypes on the antinociception induced by i.t.-administered Tyr-D-Arg-Phe-sarcosine (TAPS), a N-terminal tetrapeptide analog of dermorphin, was determined in mice tail-flick test. Intrathecal administration of TAPS produced the marked inhibition of the tail-flick response in a dose-dependent manner. The antinociception induced by TAPS was completely eliminated by i.t.-co-administration of Tyr-D-Pro-Phe-Phe-NH2 (D-Pro2-endomorphin-2), the mu1-opioid receptor antagonist, whereas i.t. co-treatment with Tyr-D-Pro-Trp-Phe-NH2 (D-Pro2-endomorphin-1) or Tyr-D-Pro-Trp-Gly-NH2 (D-Pro2-Tyr-W-MIF-1), the mu2-opioid receptor antagonists, did not affect the TAPS-induced antinociception. In contrast, the antinociception induced by i.t.-administered [D-Ala2,N-MePhe4,Gly-ol5]enkephalin was significantly attenuated by i.t.-co-administration of D-Pro2-endomorphin-1 or D-Pro2-Tyr-W-MIF-1, but not D-Pro2-endomorphin-2. These results suggest that TAPS may stimulate spinal mu1-opioid receptors to produce the antinociception.
Collapse
MESH Headings
- Analgesics/administration & dosage
- Analgesics/pharmacology
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Hot Temperature/adverse effects
- Hyperalgesia/etiology
- Hyperalgesia/metabolism
- Hyperalgesia/prevention & control
- Injections, Spinal
- MSH Release-Inhibiting Hormone/administration & dosage
- MSH Release-Inhibiting Hormone/analogs & derivatives
- MSH Release-Inhibiting Hormone/pharmacology
- Male
- Mice
- Oligopeptides/administration & dosage
- Oligopeptides/pharmacology
- Pain Measurement/methods
- Protein Isoforms/agonists
- Protein Isoforms/antagonists & inhibitors
- Protein Isoforms/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Spinal Cord/metabolism
Collapse
Affiliation(s)
- Hirokazu Mizoguchi
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fu CY, Yang Q, Wang KR, Kong ZQ, Chen Q, Wang R. Rat/mouse hemokinin-1, a mammalian tachykinin peptide, markedly potentiates the antinociceptive effects of morphine administered at the peripheral and supraspinal level. Behav Brain Res 2006; 170:293-301. [PMID: 16621052 DOI: 10.1016/j.bbr.2006.03.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2005] [Revised: 02/24/2006] [Accepted: 03/07/2006] [Indexed: 11/26/2022]
Abstract
Rat/mouse hemokinin 1 (r/m HK-1) is a mammalian tachykinin peptide whose biological functions are not fully understood. Our recent report showed that i.c.v. administration of r/m HK-1 could produce dose- and time-related antinociceptive effect at nanomole concentration, and naloxone significantly antagonized this effect. Thus, we provide indirect evidence favoring a role of NK1 supraspinal receptors in the inhibitory control of descending pain pathways, a role that seems to partially involve the activation of the endogenous opioid systems. Based on this report, the present study was conducted to further investigate the direct functional interaction between supraspinal tachykinin (r/m HK-1) and opioid systems. The results demonstrate that i.c.v. administration of r/m HK-1 (5 nmol/kg) could significantly potentiate the antinociceptive effects of morphine which was injected at peripheral and supraspinal level. These antinociceptive effects were blocked by prior treatment with the classical opioid receptors antagonist naloxone, indicating that the potentiated analgesic response is mediated by opioid-responsive neurons. Consistent with previous biochemical data, a likely mechanism underlying the peptide-mediated enhancement of opioid analgesia may center on the ability of r/m HK-1 to release endogenous opioid peptides. We suggest that there may be a cascade amplification mechanism in pain modulation when the two agents were co-administrated. The synergistic analgesic relationship of morphine and r/m HK-1 established here supports the hypothesis that supraspinal tachykinin and peripheral and central opioid systems have a direct functional interaction in the modulation of local nociceptive responses.
Collapse
Affiliation(s)
- Cai-Yun Fu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tian Shui South Road, Lanzhou 730000, People's Republic of China
| | | | | | | | | | | |
Collapse
|
38
|
Watanabe H, Nakayama D, Yuhki M, Sawai T, Sakurada W, Katsuyama S, Hayashi T, Watanabe C, Mizoguchi H, Fujimura T, Sakurada T, Sakurada S. Differential inhibitory effects of mu-opioids on substance P- and capsaicin-induced nociceptive behavior in mice. Peptides 2006; 27:760-8. [PMID: 16226344 DOI: 10.1016/j.peptides.2005.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2005] [Revised: 08/02/2005] [Accepted: 08/22/2005] [Indexed: 11/22/2022]
Abstract
The antinociceptive mechanisms of the selective mu-opioid receptor agonists [D-Ala2,NMePhe4,Gly(ol)5]enkephalin (DAMGO), H-Tyr-D-Arg-Phe-beta-Ala-OH (TAPA) or H-Tyr-D-Arg-Phe-beta-Ala-NH2 (TAPA-NH2) against substance P (SP)- or capsaicin-elicited nociceptive behaviors was investigated in mice. DAMGO, TAPA or TAPA-NH2 given intrathecally inhibited the nociceptive behaviors elicited by intrathecally administered SP or capsaicin, and these antinociceptive effects were completely eliminated by intrathecal co-administration with D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP), a selective mu-opioid receptor antagonist. Pretreatment subcutaneously with naloxonazine, a selective mu1-opioid receptor antagonist, partially attenuated the antinociceptive effect of TAPA-NH2, but not DAMGO and TAPA, against SP. However, the antinociception induced by TAPA, but not DAMGO and TAPA-NH2, against capsaicin was significantly inhibited by naloxonazine. On the other hand, co-administration intrathecally with Tyr-D-Pro-Trp-Gly-NH2 (D-Pro2-Tyr-W-MIF-1), a selective mu2-opioid receptor antagonist, significantly attenuated the antinociceptive effects of DAMGO, but not TAPA and TAPA-NH2, against capsaicin, while the antinociceptions induced by three opioid peptides against SP were significantly inhibited by D-Pro2-Tyr-W-MIF-1. These results suggest that differential inhibitory mechanisms on pre- and postsynaptic sites in the spinal cord contribute to the antinociceptive effects of the three mu-opioid peptides.
Collapse
Affiliation(s)
- Hiroyuki Watanabe
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Brumovsky P, Hofstetter C, Olson L, Ohning G, Villar M, Hökfelt T. The neuropeptide tyrosine Y1R is expressed in interneurons and projection neurons in the dorsal horn and area X of the rat spinal cord. Neuroscience 2006; 138:1361-76. [PMID: 16448775 DOI: 10.1016/j.neuroscience.2005.11.069] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Revised: 11/22/2005] [Accepted: 11/30/2005] [Indexed: 11/16/2022]
Abstract
The localization of the neuropeptide tyrosine Y1 receptor was studied with immunohistochemistry in parasagittal and transverse, free-floating sections of the rat lumbar spinal cord. At least seven distinct Y1 receptor-positive populations could tentatively be recognized: Type 1) abundant small, fusiform Y1 receptor-positive neurons in laminae I-II, producing a profuse neuropil; Type 2) Y1 receptor-positive projection neurons in lamina I; Type 3) small Y1 receptor-positive neurons in lamina III, similar to Type 1 neurons, but less densely packed; Type 4) a number of large, multipolar Y1 receptor-positive neurons in the border area between laminae III-IV, with dendrites projecting toward laminae I-II; Type 5) a considerable number of large, multipolar Y1 receptor-positive neurons in laminae V-VI; Type 6) many large Y1 receptor-positive neurons around the central canal (area X); and Type 7) a small number of large Y1 receptor-positive neurons in the medial aspect of the ventral horns (lamina VIII). Many of the neurons present in laminae V-VI and area X produce craniocaudal processes extending for several hundred micrometers. Retrograde tracing using cholera toxin B subunit injected at the 9th thoracic spinal cord level shows that several Type 5 neurons in laminae V-VI, and at least a few Type 2 in lamina I and Type 6 in area X have projections extending to the lower segments of the thoracic spinal cord (and perhaps to supraspinal levels). The present results define distinct subpopulations of neuropeptide tyrosine-sensitive neurons, localized in superficial and deep layers of the dorsal, in the ventral horns and in area X. The lamina II neurons express somatostatin [The neuropeptide Y Y1 receptor is a somatic receptor on dorsal root ganglion neurons and a postsynaptic receptor on somatostatin dorsal horn neurons. Eur J Neurosci 11:2211-2225] and are presumably glutamatergic [Todd AJ, Hughes DI, Polgar E, Nagy GG, Mackie M, Ottersen OP, Maxwell DJ (2003) The expression of vesicular glutamate transporters VGLUT1 and VGLUT2 in neurochemically defined axonal populations in the rat spinal cord with emphasis on the dorsal horn. Eur J Neurosci 17:13-27], that is they are excitatory interneurons under a Y1 receptor-mediated inhibitory influence. The remaining Y1 receptor-positive spinal neurons need to be phenotyped, for example if the large Y1 receptor-positive laminae III-IV neurons (Type 5) are identical to the neurokinin (NK)1R-positive neurons previously shown to receive neuropeptide tyrosine positive dendritic contacts [Polgár E, Shehab SA, Watt C, Todd AJ (1999) GABAergic neurons that contain neuropeptide Y selectively target cells with the NK1 receptor in laminae III and IV of the rat spinal cord. J Neurosci 19:2637-2646]. If so, neuropeptide tyrosine could have an antinociceptive action not only via Y1 receptor-positive interneurons (Type 1) but also projection neurons. The present results show neuropeptide tyrosine-sensitive neuron populations virtually in all parts of the lumbar spinal cord, suggesting a role for neuropeptide tyrosine signaling in many spinal functions, including pain.
Collapse
Affiliation(s)
- P Brumovsky
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, B2:5, S-171 77 Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
40
|
Fu CY, Kong ZQ, Wang KR, Yang Q, Zhai K, Chen Q, Wang R. Effects and mechanisms of supraspinal administration of rat/mouse hemokinin-1, a mammalian tachykinin peptide, on nociception in mice. Brain Res 2005; 1056:51-8. [PMID: 16102736 DOI: 10.1016/j.brainres.2005.07.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 07/13/2005] [Accepted: 07/13/2005] [Indexed: 10/25/2022]
Abstract
Rat/mouse hemokinin 1 (r/m HK-1) is a novel tachykinin peptide whose biological functions are not fully understood. This work was designed to observe the effects of r/m HK-1 in pain modulation at supraspinal level in mice using tail-flick test. Intracerebroventricular (i.c.v.) administration of r/m HK-1 (0.1, 0.3, 1, 3 nmol/mouse) dose-dependently induced potent analgesic effect (ED(50) = 0.2877 nmol/mouse). When r/m HK-1 co-injected (i.c.v.) with SR140333 (a selective NK(1) receptor antagonist), SR140333 could fully antagonize the analgesic effect of r/m HK-1. The maximal analgesic effect of r/m HK-1 (3 nmol/mouse) could also be reversed by naloxone (i.p., 2 mg/kg). However, i.c.v. low dose administration of r/m HK-1 (10, 3, 1 pmol/mouse) induced hyperalgesia with a "U" shape curve, which means that the maximal hyperalgesic effect appeared at 3 pmol/mouse, and this effect of r/m HK-1 could also be fully blocked by SR140333. Interestingly, [Nphe(1)]NC(1-13)NH(2), a selective opioid receptor like-1 (ORL-1) receptor antagonist, could fully reverse the maximal hyperalgesic effect of r/m HK-1 (3 pmol/mouse). In addition, when r/m HK-1 co-injected (i.c.v.) with SR48968 (a selective NK(2) receptor antagonist), SR48968 could hardly affect the nociceptive effects of r/m HK-1 either at nanomole concentration or at picomole concentration. These findings suggested that r/m HK-1 might play an important role in pain modulation at supraspinal level in mice and these effects were first elicited through the activation of NK(1) receptor, subsequently, whether activation of the classical opioid receptor or the ORL1 receptor depending on the dose of i.c.v. administration of r/m HK-1.
Collapse
Affiliation(s)
- Cai-Yun Fu
- Department of Biochemistry and Molecular Biology, School of Life Science, Lanzhou University, 222 Tian Shui South Road, Lanzhou 730000, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
41
|
Mitchell JL, Silverman MB, Aicher SA. Rat trigeminal lamina I neurons that project to thalamic or parabrachial nuclei contain the mu-opioid receptor. Neuroscience 2005; 128:571-82. [PMID: 15381286 DOI: 10.1016/j.neuroscience.2004.07.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2004] [Indexed: 10/26/2022]
Abstract
Ligands of the mu-opioid receptor are known to inhibit nociceptive transmission in the dorsal horn, yet the cellular site(s) of action for this inhibition remain to be fully elucidated. Neurons located in lamina I of the dorsal horn are involved in distinct aspects of nociceptive transmission. Neurons projecting to the thalamus are thought to be involved in sensory-discriminative aspects of pain perception, while neurons projecting to the parabrachial nucleus are thought to be important for emotional and/or autonomic responses to noxious stimuli. The present study examined these two populations of lamina I projection neurons in the trigeminal dorsal horn to determine if the mu-opioid receptor protein (MOR1) is differentially located in these populations of neurons. Lamina I projection neurons were identified using the retrograde tracer FluoroGold (FGold). FGold was injected into either the contralateral thalamus (ventral posterolateral (VPM)/ventral posterolateral (VPL) thalamic region) or into the ipsilateral parabrachial nuclei. The distribution of MOR1 in these neurons was determined using immunocytochemistry. The distribution of MOR1-ir within these two populations of lamina I projection neurons was examined by both confocal and electron microscopy. We found that both populations of projection neurons contained MOR1. Immunogold analyses revealed the presence of MOR1-ir at membrane sites and within the cytoplasm of these neurons. Cytoplasmic receptor labeling may represent sites of synthesis, recycling or reserve populations of receptors. MOR1 was primarily found in the somata and proximal dendrites of projection neurons. In addition, these neurons rarely received synaptic input from MOR1-containing axon terminals. These results indicate that lamina I neurons in trigeminal dorsal horn that project to the thalamic and parabrachial nuclei contain MOR1 and are likely sites of action for MOR ligands that modulate sensory and/or autonomic aspects of pain transmission in the trigeminal dorsal horn.
Collapse
Affiliation(s)
- J L Mitchell
- Neurological Sciences Institute, Oregon Health and Science University, 505 Northwest 185(th) Avenue, Beaverton, OR 97006, USA
| | | | | |
Collapse
|
42
|
Silverman MB, Hermes SM, Zadina JE, Aicher SA. Mu-opioid receptor is present in dendritic targets of Endomorphin-2 axon terminals in the nuclei of the solitary tract. Neuroscience 2005; 135:887-96. [PMID: 16154285 DOI: 10.1016/j.neuroscience.2005.06.072] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2005] [Revised: 06/23/2005] [Accepted: 06/27/2005] [Indexed: 11/26/2022]
Abstract
Endomorphins represent a group of endogenous opioid peptides with high affinity for the mu-opioid receptor. In the brainstem, Endomorphin-2 is found in trigeminal dorsal horn and the nuclei of the solitary tract, suggesting its presence in both nociceptive and visceral primary afferents. If Endomorphin-2 were an endogenous ligand for the mu-opioid receptor, we would expect to find the receptor at cellular sites in close association with the peptide. We used dual-labeling immunocytochemistry combined with electron microscopy to examine interactions between Endomorphin-2-immunoreactive and mu-opioid receptor-immunoreactive profiles within the nuclei of the solitary tract in the rat. Endomorphin-2-immunoreactivity was found primarily in unmyelinated axons and axon terminals in nuclei of the solitary tract and the majority of these terminals contained dense core vesicles. Endomorphin-2-immunoreactive axon terminals often formed asymmetric synapses with dendritic spines lacking mu-opioid receptor-immunoreactivity, but mu-opioid receptor-immunoreactivity was found in many of the larger dendritic targets of Endomorphin-2-immunoreactive terminals. Thus, mu-opioid receptor-immunoreactivity was found in the postsynaptic targets of Endomorphin-2-immunoreactive axon terminals, consistent with the hypothesis that Endomorphin-2 is an endogenous ligand for this receptor within the nuclei of the solitary tract. A small number of Endomorphin-2-immunoreactive somata, dendrites, and axon terminals also contained mu-opioid receptor-immunoreactivity. Cells that contain both the opioid peptide and its receptor may be a substrate for potential autoregulation of nuclei of the solitary tract neurons by opioid ligands. Finally, using tract tracing and confocal microscopy, we found Endomorphin-2-immunoreactivity in a subset of vagal afferents. Together these findings support the hypothesis that Endomorphin-2 is a ligand for the mu-opioid receptor within nuclei of the solitary tract and that the peptide is at least partially derived from primary visceral afferents.
Collapse
Affiliation(s)
- M B Silverman
- Oregon Health & Science University, Neurological Sciences Institute, 505 Northwest 185th Avenue, Beaverton, OR 97006, USA
| | | | | | | |
Collapse
|
43
|
Gamboa-Esteves FO, McWilliam PN, Batten TFC. Substance P (NK1) and somatostatin (sst2A) receptor immunoreactivity in NTS-projecting rat dorsal horn neurones activated by nociceptive afferent input. J Chem Neuroanat 2004; 27:251-66. [PMID: 15261332 DOI: 10.1016/j.jchemneu.2004.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2003] [Revised: 02/17/2004] [Accepted: 04/06/2004] [Indexed: 10/26/2022]
Abstract
Spinal neurones that receive inputs from primary afferent fibres and have axons projecting supraspinally to the medulla oblongata may represent a pathway through which nociceptive and non-nociceptive peripheral stimuli are able to modulate cardiorespiratory reflexes. Expression of the neurokinin-1 (NK1) receptor is believed to be an indicator of lamina I cells that receive nociceptive inputs from substance P releasing afferents, and similarly, sst2A receptor expression may be a marker for neurones receiving somatostatinergic inputs. In this study, immunoreactivity for these two receptors was investigated in rat spinal neurones retrogradely labelled by injections of cholera toxin B or Fluorogold into the nucleus of the solitary tract (NTS). In addition, nociceptive activation of these labelled cells was studied by immunodetection of Fos protein in response to cutaneous and visceral noxious chemical stimuli. NK1 and sst2A receptors in lamina I were localised to mainly separate populations of retrogradely labelled cells with fusiform, flattened and pyramidal morphologies. Examples of projection neurones expressing both receptors were, however observed. With visceral stimulation, many retrogradely labelled cells expressing c-fos were immunoreactive for the NK1 receptor, and a smaller population was sst2A positive. In contrast, with cutaneous stimulation, only NK1 positive retrogradely labelled cells showed c-fos expression. These data provide evidence that lamina I neurones receiving noxious cutaneous and visceral stimuli via NK1 receptor activation project to NTS and so may be involved in coordinating nociceptive and cardiorespiratory responses. Moreover, a subpopulation of projection neurones that respond to visceral stimuli may receive somatostatinergic inputs of peripheral, local or supraspinal origins.
Collapse
Affiliation(s)
- Filomena O Gamboa-Esteves
- Institute for Cardiovascular Research, School of Medicine, Worsley Building, University of Leeds, Leeds LS2 9JT, UK
| | | | | |
Collapse
|
44
|
Morris R, Cheunsuang O, Stewart A, Maxwell D. Spinal dorsal horn neurone targets for nociceptive primary afferents: do single neurone morphological characteristics suggest how nociceptive information is processed at the spinal level. ACTA ACUST UNITED AC 2004; 46:173-90. [PMID: 15464206 DOI: 10.1016/j.brainresrev.2004.07.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2004] [Indexed: 01/03/2023]
Abstract
It has become increasingly clear that nociceptive information is signalled by several anatomically distinct populations of primary afferents that target different populations of neurones in the spinal cord. It is probable that these different systems all give rise to the sensation pain and hence, an understanding of their separate roles and the processes that they employ, may offer ways of selectively targeting pain arising from different causes. The review focuses on what is known of the anatomy of neurones in LI-III of the spinal dorsal horn that are implicated in nociception. The dendritic geometry and synaptic input of the large LI neurones that receive input from primary afferents containing substance P that express neurokinin 1 (NK(1)) receptors suggests that these neurones may monitor the extent of injury rather than the specific localisation of a discrete noxious stimulus. This population of neurones is also critically involved in hyperalgesia. In contrast neurones in LII with the morphology of stalked cells that receive primary afferent input from glomerular synapses may be more suitable for fine discrimination of the exact location of a noxious event such as a sting or parasite attack. The review focuses as far as possible on precisely defined anatomy in the belief that only by understanding these anatomical relationships will we eventually be able to interpret the complex processes occurring in the dorsal horn. The review attempts to be an accessible guide to a sometimes complex and highly specialised literature in this field.
Collapse
Affiliation(s)
- Richard Morris
- Department of Veterinary Preclinical Sciences, University of Liverpool, Brownlow Hill/Crown Street, Liverpool, L69 7ZJ, UK.
| | | | | | | |
Collapse
|
45
|
Doly S, Fischer J, Conrath M. The vanilloid receptor-1 (TRPV1) is expressed in some rat dorsal horn NK1 cells. Brain Res 2004; 1004:203-7. [PMID: 15033437 DOI: 10.1016/j.brainres.2004.01.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2004] [Indexed: 11/30/2022]
Abstract
The vanilloid receptor-1 (TRPV1), the capsaicin receptor, a transducer of several nociceptive stimuli, is principally expressed by nociceptive fibers that specifically target supraspinal-projecting neurons expressing the NK1 receptor. TRPV1 is also expressed by intraspinal neurons. Using double immunocytochemistry, we show that 14.2% of these TRPV1 dorsal horn cell bodies also express the NK1 receptor suggesting that endogenous vanilloids may directly modulate second-order ascending neurons.
Collapse
Affiliation(s)
- Stéphane Doly
- CNRS UMR 7101, Neurobiologie des Signaux Intercellulaires, Université Pierre et Marie Curie, 7 Quai Saint Bernard Bat A 3em, 75252 Paris, France.
| | | | | |
Collapse
|
46
|
Rosén A, Zhang YX, Lund I, Lundeberg T, Yu LC. Substance P microinjected into the periaqueductal gray matter induces antinociception and is released following morphine administration. Brain Res 2004; 1001:87-94. [PMID: 14972657 DOI: 10.1016/j.brainres.2003.11.060] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2003] [Indexed: 11/23/2022]
Abstract
The aims of the present study were to investigate, in rats, the behavioral effects of substance P (SP) microinjected into the ventrolateral periaqueductal gray (PAG) and the effects of the neurokinin 1 (NK-1) receptor antagonist [d-Arg1, d-Trp7, 9, Leu11]-substance P (Spantide). The effect of morphine administration on the release of SP in the ventrolateral PAG was also investigated using microdialysis in awake rats. SP microinjected into the ventrolateral part of the PAG induced significant increases in the hindpaw withdrawal latencies (HWLs) to thermal and mechanical stimulation as an antinociceptive response. The NK-1 receptor antagonist blocked these effects but exhibited no antinociceptive effect alone. Subcutaneous administration of morphine increased basal SP-like immunoreactivity (SP-LI) release in the microdialysate obtained from the ventrolateral PAG of freely moving rats. Our results demonstrate that SP injected into the ventrolateral PAG induces an antinociceptive effect via activation of NK-1 receptors. Morphine administered systemically induces the release of SP in the ventrolateral PAG. We suggest that an increased release of SP in the PAG may contribute to opioid antinociception.
Collapse
Affiliation(s)
- Annika Rosén
- Department of Odontology, Division of Oral and Maxillofacial Surgery, Karolinska Institutet, Huddinge Hospital, Box 4064, SE-141 04 Huddinge, Sweden.
| | | | | | | | | |
Collapse
|
47
|
Aicher SA, Goldberg A, Sharma S. Co-localization of mu opioid receptor and N-methyl-D-aspartate receptor in the trigeminal dorsal horn. THE JOURNAL OF PAIN 2003; 3:203-10. [PMID: 14622774 DOI: 10.1054/jpai.2002.123709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Antagonists acting at the N-methyl-D-aspartate (NMDA) receptor can block the development of tolerance to the analgesic effects of [mu ] opioid receptor (MOR) ligands, such as morphine, and can also enhance the analgesic efficacy of opioids. These findings have led to the hypothesis that interactions between NMDA receptor and MOR ligands may be due to the co-localization of these receptors on neurons in the dorsal horn. We used dual immunogold and immunoperoxidase immunocytochemistry for MOR1 and NMDAR1 to determine the degree of co-localization of these receptors in neurons of the trigeminal dorsal horn. By use of electron microscopy, we found that both receptors were primarily located in dendrites and to a lesser extent in perikarya, axons, axon terminals, and glia. With regard to the degree of co-localization in dendrites, 63% of MOR1-labeled dendrites also contained NMDAR1, whereas 61% of NMDAR1-labeled dendrites also contained MOR1. Most of the dual-labeled profiles (94%) were classified as dendrites, with the remainder being axons, axon terminals, or perikarya. These results suggest that direct interactions between MOR and NMDA receptor ligands are likely mediated through shared dendritic targets in the dorsal horn. Less frequently, we found evidence for modulation of afferents to MOR-containing neurons through presynaptic NMDA receptors.
Collapse
Affiliation(s)
- Sue A Aicher
- Oregon Health Sciences University, Neurological Sciences Institute, Beaverton, 97006, USA.
| | | | | |
Collapse
|
48
|
Pfeiffer M, Kirscht S, Stumm R, Koch T, Wu D, Laugsch M, Schröder H, Höllt V, Schulz S. Heterodimerization of substance P and mu-opioid receptors regulates receptor trafficking and resensitization. J Biol Chem 2003; 278:51630-7. [PMID: 14532289 DOI: 10.1074/jbc.m307095200] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The micro-opioid receptor (MOR1) and the substance P receptor (NK1) coexist and functionally interact in nociceptive brain regions; however, a molecular basis for this interaction has not been established. Using coimmunoprecipitation and bioluminescence resonance energy transfer (BRET), we show that MOR1 and NK1 can form heterodimers in HEK 293 cells coexpressing the two receptors. Although NK1-MOR1 heterodimerization did not substantially change the ligand binding and signaling properties of these receptors, it dramatically altered their internalization and resensitization profile. Exposure of the NK1-MOR1 heterodimer to the MOR1-selective ligand [D-Ala2,Me-Phe4,Gly5-ol]enkephalin (DAMGO) promoted cross-phosphorylation and cointernalization of the NK1 receptor. Conversely, exposure of the NK1-MOR1 heterodimer to the NK1-selective ligand substance P (SP) promoted cross-phosphorylation and cointernalization of the MOR1 receptor. In cells expressing MOR1 alone, beta-arrestin directs the receptors to clathrin-coated pits, but does not internalize with the receptor. In cells expressing NK1 alone, beta-arrestin internalizes with the receptor into endosomes. Interestingly, in cells coexpressing MOR1 and NK1 both DAMGO and SP induced the recruitment of beta-arrestin to the plasma membrane and cointernalization of NK1-MOR1 heterodimers with beta-arrestin into the same endosomal compartment. Consequently, resensitization of MOR1-dependent receptor functions was severely delayed in coexpressing cells as compared with cells expressing MOR1 alone. Together, our findings indicate that MOR1 by virtue of its physical interaction with NK1 is sequestered via an endocytotic pathway with delayed recycling and resensitization kinetics.
Collapse
Affiliation(s)
- Manuela Pfeiffer
- Department of Pharmacology and Toxicology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vierck CJ, Kline RH, Wiley RG. Intrathecal substance p-saporin attenuates operant escape from nociceptive thermal stimuli. Neuroscience 2003; 119:223-32. [PMID: 12763083 DOI: 10.1016/s0306-4522(03)00125-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Destruction of neurons in the superficial dorsal horn that express substance P receptor (NK-1R) has been reported to block development of behavioral hypersensitivity following peripheral sensitization of nociceptors. Baseline sensitivity was not altered in these rat models that assessed innate reflex responses (i.e. hind-paw withdrawal to thermal or mechanical stimulation). In the present study, we evaluated effects of intrathecal substance P-saporin (SP-sap), a toxin selective for cells expressing NK-1R, on operant escape responses of rats to thermal stimulation. For comparison, lick/guard reflex testing was performed. Injection of a modest dose (175 ng) of SP-sap into the lumbar subarachnoid space produced a partial loss of lamina I/II NK-1R-expressing dorsal horn neurons but did not affect NK-1R-expressing neurons in deeper laminae. Lick/guard responses to 0.3, 44 or 47 degrees C were not affected after SP-sap treatment, but escape responses to these temperatures were significantly attenuated. Three hours after application of mustard oil to the dorsal surface of both hind paws, escape from 44 degrees C was enhanced for controls but not SP-sap-treated rats. Lick/guard responses were enhanced by mustard oil for both SP-sap and control animals. Administration of morphine (1.0 mg/kg, s.c.) before testing decreased escape responding at 47 degrees C for both controls and SP-sap rats. Thus, partial loss of NK-1R-expressing neurons in the superficial dorsal horn attenuated thermal nociceptive sensitivity and prevented secondary hyperalgesia when studied with an operant algesia assay, in contrast to innate reflexes which were less sensitive to modification by intrathecal SP-sap.
Collapse
Affiliation(s)
- C J Vierck
- Department of Neuroscience and McKnight Brain Institute, University of Florida College of Medicine, P.O. Box 100244, Gainesville, FL 32610, USA.
| | | | | |
Collapse
|
50
|
Aicher SA, Mitchell JL, Swanson KC, Zadina JE. Endomorphin-2 axon terminals contact mu-opioid receptor-containing dendrites in trigeminal dorsal horn. Brain Res 2003; 977:190-8. [PMID: 12834879 DOI: 10.1016/s0006-8993(03)02678-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The endomorphins represent a novel group of endogenous opioid peptides that have high affinity for the mu-opioid receptor (MOR1). Endomorphin-2 is present in high density in the spinal and trigeminal dorsal horns and is localized to primary afferents. If endomorphin-2 were an endogenous ligand for the MOR1, we would expect to find the receptor at cellular sites in close association with the peptide. We used dual-labeling immunocytochemical methods combined with electron microscopy to determine if a cellular substrate exists for functional interactions between endomorphin-2 and MOR1. We confirmed the localization of endomorphin-2 to unmyelinated axons and axon terminals in the trigeminal dorsal horn. A small proportion of these endomorphin-2 axons contained MOR1, but many of the dendritic targets of endomorphin-2 terminals contained MOR1. Consistent with previous studies, endomorphin-2 was contained primarily in dense core vesicles and MOR1 was located primarily at non-synaptic sites. These morphological characteristics are consistent with the hypothesis that peptides are released extra-synaptically and their receptors may be located at sites distal to the synaptic junction. These anatomical data support the hypothesis that endomorphin-2 is a ligand for MORs in the trigeminal dorsal horn, particularly at postsynaptic sites.
Collapse
Affiliation(s)
- Sue A Aicher
- Neurological Sciences Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
| | | | | | | |
Collapse
|