1
|
Micheli L, Nobili S, Lucarini E, Toti A, Margiotta F, Ciampi C, Venturi D, Di Cesare Mannelli L, Ghelardini C. New insights in the mechanisms of opioid analgesia and tolerance: Ultramicronized palmitoylethanolamide down-modulates vascular endothelial growth factor-A in the nervous system. Pharmacol Res 2024; 209:107472. [PMID: 39448045 DOI: 10.1016/j.phrs.2024.107472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Growing evidence suggests that opioid analgesics modulate angiogenesis during pathophysiological processes. Vascular endothelial growth factor-A (VEGF-A) was recently proposed to be involved in pain development. To date, no anti-angiogenic drug is used for pain management. When administered in a bioavailable formulation, (i.e., ultramicronized) N-palmitoylethanolamine (PEA) delays the onset of morphine tolerance, improves morphine analgesic activity and reduces angiogenesis in in vivo models. This study aimed at investigating whether VEGF-A is involved in PEA-induced delay of morphine tolerance. The anti-VEGF-A monoclonal antibody bevacizumab was used as a reference drug. Preemptive and concomitant treatment with ultramicronized PEA delayed morphine tolerance and potentiated the analgesic effect of morphine, while counteracting morphine-induced increase of VEGF-A in the nervous system. Similar results were obtained when bevacizumab was administered together with morphine. Of note, bevacizumab showed an analgesic effect per se. In equianalgesic treatment regimens (obtained through increasing morphine doses and associating PEA), PEA resulted in lower expression of VEGF-A in dorsal root ganglia (DRG) and spinal cord compared to morphine alone. Similar results were observed for plasma levels of the soluble VEGF receptor 1 (sFLT-1). Moreover, in morphine-treated animals, two pain-related genes (i.e., Serpina3n and Eaat2) showed a more than 3-fold increase in their expression at spinal cord and DRG level, with the increase being significantly counteracted by PEA treatment. This study supports the hypothesis that the effects of PEA on morphine analgesia and tolerance may be mediated by the down-modulation of VEGF-A and sFLT-1 in the nervous system and plasma, respectively.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy.
| | - Stefania Nobili
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Francesco Margiotta
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Daniel Venturi
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| |
Collapse
|
2
|
Jensen KL, Christensen NR, Goddard CM, Jager SE, Noes-Holt G, Kanneworff IB, Jakobsen A, Jiménez-Fernández L, Peck EG, Sivertsen L, Comaposada Baro R, Houser GA, Mayer FP, Diaz-delCastillo M, Topp ML, Hopkins C, Thomsen CD, Soltan ABI, Tidemand FG, Arleth L, Heegaard AM, Sørensen AT, Madsen KL. Peripherally restricted PICK1 inhibitor mPD5 ameliorates pain behaviors in murine inflammatory and neuropathic pain models. JCI Insight 2024; 9:e170976. [PMID: 39287978 PMCID: PMC11530130 DOI: 10.1172/jci.insight.170976] [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: 03/29/2023] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Chronic pain is a complex, debilitating, and escalating health problem worldwide, impacting 1 in 5 adults. Current treatment is compromised by dose-limiting side effects, including high abuse liability, loss of ability to function socially and professionally, fatigue, drowsiness, and apathy. PICK1 has emerged as a promising target for the treatment of chronic pain conditions. Here, we developed and characterized a cell-permeable fatty acid-conjugated bivalent peptide inhibitor of PICK1 and assessed its effects on acute and chronic pain. The myristoylated PICK1 inhibitor, myr-NPEG4-(HWLKV)2 (mPD5), self-assembled into core-shell micelles that provided favorable pharmacodynamic properties and relieved evoked mechanical and thermal hypersensitivity as well as ongoing hypersensitivity and anxiodepressive symptoms in mouse models of neuropathic and inflammatory pain following subcutaneous administration. No overt side effects were associated with mPD5 administration, and it had no effect on acute nociception. Finally, neuropathic pain was relieved far into the chronic phase (18 weeks after spared nerve injury surgery) and while the effect of a single injection ceased after a few hours, repeated administration provided pain relief lasting up to 20 hours after the last injection.
Collapse
Affiliation(s)
| | - Nikolaj Riis Christensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, and
| | | | - Sara Elgaard Jager
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
| | - Gith Noes-Holt
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
| | - Ida Buur Kanneworff
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Jakobsen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
| | | | - Emily G. Peck
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Line Sivertsen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
| | | | - Grace Anne Houser
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
| | - Felix Paul Mayer
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
| | - Marta Diaz-delCastillo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Løth Topp
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience
| | - Chelsea Hopkins
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Dubgaard Thomsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ahmed Barakat Ibrahim Soltan
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederik Grønbæk Tidemand
- X-ray and Neutron Science, Niels Bohr Institute, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Lise Arleth
- X-ray and Neutron Science, Niels Bohr Institute, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
3
|
Ciapała K, Mika J. Advances in Neuropathic Pain Research: Selected Intracellular Factors as Potential Targets for Multidirectional Analgesics. Pharmaceuticals (Basel) 2023; 16:1624. [PMID: 38004489 PMCID: PMC10675751 DOI: 10.3390/ph16111624] [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: 10/26/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Neuropathic pain is a complex and debilitating condition that affects millions of people worldwide. Unlike acute pain, which is short-term and starts suddenly in response to an injury, neuropathic pain arises from somatosensory nervous system damage or disease, is usually chronic, and makes every day functioning difficult, substantially reducing quality of life. The main reason for the lack of effective pharmacotherapies for neuropathic pain is its diverse etiology and the complex, still poorly understood, pathophysiological mechanism of its progression. Numerous experimental studies, including ours, conducted over the last several decades have shown that the development of neuropathic pain is based on disturbances in cell activity, imbalances in the production of pronociceptive factors, and changes in signaling pathways such as p38MAPK, ERK, JNK, NF-κB, PI3K, and NRF2, which could become important targets for pharmacotherapy in the future. Despite the availability of many different analgesics, relieving neuropathic pain is still extremely difficult and requires a multidirectional, individual approach. We would like to point out that an increasing amount of data indicates that nonselective compounds directed at more than one molecular target exert promising analgesic effects. In our review, we characterize four substances (minocycline, astaxanthin, fisetin, and peimine) with analgesic properties that result from a wide spectrum of actions, including the modulation of MAPKs and other factors. We would like to draw attention to these selected substances since, in preclinical studies, they show suitable analgesic properties in models of neuropathy of various etiologies, and, importantly, some are already used as dietary supplements; for example, astaxanthin and fisetin protect against oxidative stress and have anti-inflammatory properties. It is worth emphasizing that the results of behavioral tests also indicate their usefulness when combined with opioids, the effectiveness of which decreases when neuropathy develops. Moreover, these substances appear to have additional, beneficial properties for the treatment of diseases that frequently co-occur with neuropathic pain. Therefore, these substances provide hope for the development of modern pharmacological tools to not only treat symptoms but also restore the proper functioning of the human body.
Collapse
Affiliation(s)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Str., 31-343 Kraków, Poland;
| |
Collapse
|
4
|
Xi KW, Chen DD, Geng X, Bian Y, Wang MX, Bian H. Calcium/calmodulin-dependent protein kinase II is involved in the transmission and regulation of nociception in naïve and morphine-tolerant rat nucleus accumbens. Korean J Pain 2023; 36:163-172. [PMID: 36941088 PMCID: PMC10043793 DOI: 10.3344/kjp.22372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 03/23/2023] Open
Abstract
Background Synaptic plasticity contributes to nociceptive signal transmission and modulation, with calcium/calmodulin-dependent protein kinase II (CaMK II) playing a fundamental role in neural plasticity. This research was conducted to investigate the role of CaMK II in the transmission and regulation of nociceptive information within the nucleus accumbens (NAc) of naïve and morphine-tolerant rats. Methods Randall Selitto and hot-plate tests were utilized to measure the hindpaw withdrawal latencies (HWLs) in response to noxious mechanical and thermal stimuli. To induce chronic morphine tolerance, rats received intraperitoneal morphine injection twice per day for seven days. CaMK II expression and activity were assessed using western blotting. Results Intra-NAc microinjection of autocamtide-2-related inhibitory peptide (AIP) induced an increase in HWLs in naïve rats in response to noxious thermal and mechanical stimuli. Moreover, the expression of the phosphorylated CaMK II (p-CaMK II) was significantly decreased as determined by western blotting. Chronic intraperitoneal injection of morphine resulted in significant morphine tolerance in rats on Day 7, and an increase of p-CaMK II expression in NAc in morphine-tolerant rats was observed. Furthermore, intra-NAc administration of AIP elicited significant antinociceptive responses in morphine-tolerant rats. In addition, compared with naïve rats, AIP induced stronger thermal antinociceptive effects of the same dose in rats exhibiting morphine tolerance. Conclusions This study shows that CaMK II in the NAc is involved in the transmission and regulation of nociception in naïve and morphine-tolerant rats.
Collapse
Affiliation(s)
- Kai Wen Xi
- Department of Physiology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
- Second Department of Neurosurgery, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
- Department of Cerebrovascular Surgery, Xinyu People’s Hospital, Xinyu, Jiangxi, China
| | - De Duo Chen
- Department of Physiology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xin Geng
- Second Department of Neurosurgery, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Yan Bian
- Department of Oncology, The Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Min Xin Wang
- Department of Physiology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Hui Bian
- Department of Physiology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
5
|
Fu X, Zhang Y. Research progress of p38 as a new therapeutic target against morphine tolerance and the current status of therapy of morphine tolerance. J Drug Target 2023; 31:152-165. [PMID: 36264036 DOI: 10.1080/1061186x.2022.2138895] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
With the development of the medical industry, new painkillers continue to appear in people's field of vision, but so far no painkiller can replace morphine. While morphine has a strong analgesic effect, it is also easy to produce pain sensitivity and tolerance. Due to the great inter-individual differences in patient responses, there are few clear instructions on how to optimise morphine administration regimens, which complicates clinicians' treatment strategies and limits the effectiveness of morphine in long-term pain therapy. P38MAPK is a key member of the MAPK family. Across recent years, it has been discovered that p38MAPK rises dramatically in a wide range of morphine tolerance animal models. Morphine tolerance can be reduced or reversed by inhibiting p38MAPK. However, the role and specific mechanism of p38MAPK are not clear. In this review, we synthesise the relevant findings, highlight the function and potential mechanism of p38MAPK in morphine tolerance, as well as the present status and efficacy of morphine tolerance therapy, and underline the future promise of p38MAPK targeted morphine tolerance treatment.
Collapse
Affiliation(s)
- Xiao Fu
- Inner Mongolia Medical University, Hohhot, China
| | - Yanhong Zhang
- Department of Anesthesiology, People's Hospital Affiliated to Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
6
|
Wolińska R, Kleczkowska P, de Cordé-Skurska A, Poznański P, Sacharczuk M, Mika J, Bujalska-Zadrożny M. Nitric oxide modulates tapentadol antinociceptive tolerance and physical dependence. Eur J Pharmacol 2021; 907:174245. [PMID: 34126091 DOI: 10.1016/j.ejphar.2021.174245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
Tapentadol, an analgesic with a dual mechanism of action, involving both μ-opioid receptor agonism and noradrenaline reuptake inhibition (MOP-NRI), was designed for the treatment of moderate to severe pain. However, the widely acknowledged risk of analgesic tolerance and development of physical dependence following sustained opioid use may hinder their effectiveness. One of the possible mechanisms behind these phenomena are alterations in nitric oxide synthase (NOS) system activity. The aim of the study was to investigate the tolerance and dependence potential of tapentadol in rodent models and to evaluate the possible role of nitric oxide (NO) in these processes. Our study showed that chronic tapentadol treatment resulted in tolerance to its antinociceptive effects to an extent similar to tramadol, but much less than morphine. A single injection of a non-selective NOS inhibitor, NG-nitro-L-arginine (L-NOArg), reversed the tapentadol tolerance. In dependence studies, repeated administration of L-NOArg attenuated naloxone-precipitated withdrawal in tapentadol-treated mice, whereas a single injection of L-NOArg was ineffective. Biochemical analysis revealed that tapentadol decreased nNOS protein levels in the dorsal root ganglia of rats following 31 days of treatment, while no significant changes were found in iNOS and eNOS protein expression. Moreover, pre-treatment with L-NOArg augmented tapentadol antinociception in an opioid- and α2-adrenoceptor-dependent manner. In conclusion, our data suggest that the NOS system plays an important role in the attenuation of tapentadol-induced tolerance and withdrawal. Thus, inhibition of NOS activity can serve as a promising treatment option for long-term tapentadol use by extending its effectiveness and improving the side-effects profile.
Collapse
Affiliation(s)
- Renata Wolińska
- Department of Pharmacodynamics, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1b Banacha Street, 01-793 Warsaw, Poland.
| | - Patrycja Kleczkowska
- Department of Pharmacodynamics, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1b Banacha Street, 01-793 Warsaw, Poland; Military Institute of Hygiene and Epidemiology, 4 Kozielska Street, 01-163 Warsaw, Poland
| | - Anna de Cordé-Skurska
- Department of Pharmacodynamics, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1b Banacha Street, 01-793 Warsaw, Poland
| | - Piotr Poznański
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences in Jastrzebiec, Postepu 36A Street, 05-552 Magdalenka, Poland
| | - Mariusz Sacharczuk
- Department of Pharmacodynamics, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1b Banacha Street, 01-793 Warsaw, Poland; Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences in Jastrzebiec, Postepu 36A Street, 05-552 Magdalenka, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - Magdalena Bujalska-Zadrożny
- Department of Pharmacodynamics, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1b Banacha Street, 01-793 Warsaw, Poland
| |
Collapse
|
7
|
Wang T, Zhu X, Yi H, Gu J, Liu S, Izenwasser S, Lemmon VP, Roy S, Hao S. Viral vector-mediated gene therapy for opioid use disorders. Exp Neurol 2021; 341:113710. [PMID: 33781732 DOI: 10.1016/j.expneurol.2021.113710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
Chronic exposure to opioids typically results in adverse consequences. Opioid use disorder (OUD) is a disease of the CNS with behavioral, psychological, neurobiological, and medical manifestations. OUD induces a variety of changes of neurotransmitters/neuropeptides in the nervous system. Existing pharmacotherapy, such as opioid maintenance therapy (OMT) is the mainstay for the treatment of OUD, however, current opioid replacement therapy is far from effective for the majority of patients. Pharmacological therapy for OUD has been challenging for many reasons including debilitating side-effects. Therefore, developing an effective, non-pharmacological approach would be a critical advancement in improving and expanding treatment for OUD. Viral vector mediated gene therapy provides a potential new approach for treating opioid abused patients. Gene therapy can supply targeting gene products directly linked to the mechanisms of OUD to restore neurotransmitter and/or neuropeptides imbalance, and avoid the off-target effects of systemic administration of drugs. The most commonly used viral vectors in rodent studies of treatment of opioid-used disorder are based on recombinant adenovirus (AV), adeno-associated virus (AAV), lentiviral (LV) vectors, and herpes simplex virus (HSV) vectors. In this review, we will focus on the recent progress of viral vector mediated gene therapy in OUD, especially morphine tolerance and withdrawal.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Xun Zhu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hyun Yi
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Jun Gu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shue Liu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sari Izenwasser
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Vance P Lemmon
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shuanglin Hao
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America.
| |
Collapse
|
8
|
Kashiwagi Y, Yi H, Liu S, Takahashi K, Hayashi K, Ikegami D, Zhu X, Gu J, Hao S. Mitochondrial biogenesis factor PGC-1α suppresses spinal morphine tolerance by reducing mitochondrial superoxide. Exp Neurol 2021; 339:113622. [PMID: 33516729 DOI: 10.1016/j.expneurol.2021.113622] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 11/15/2022]
Abstract
Opioid use disorders (OUDs) have reached an epidemic level in the United States. The opioid epidemic involves illicit opioid use, prescription opioids for analgesia, counterfeit opioids, new psychoactive substances, and diverted opioids. Opioids remain the last option for the treatment of intractable clinical pain, but chronic use of opioids are limited in part due to antinociceptive/analgesic tolerance. Peroxisome proliferator-activated receptor (PPAR)-gamma coactivator-1alpha (PGC-1α), a mitochondrial biogenesis factor can reduce toxic reactive oxygen species (ROS) that play a role in morphine tolerance (MT). Decreased PGC-1α expression has been shown to contribute to various metabolic disorders or neurodegeneration diseases through increasing ROS. We examined the relationship of PGC-1α and ROS in MT. To induce MT, adult Sprague-Dawley rats received intrathecal morphine for 7 days. Mechanical threshold was measured using the von Frey test and thermal latency was examined using the heat plate test. Expression of PGC-1α in the spinal cord dorsal horn (SCDH) was examined using RT-PCR and western blots. Mitochondrial superoxide was detected using MitoSox Red, a mitochondrial superoxide indicator. The antinociceptive effect of recombinant PGC-1α (rPGC-1α) or Mito-Tempol (a mitochondria-targeted superoxide scavenger) was determined using the von Frey test and hot plate test. Furthermore, we examined the effect of rPGC-1α on mitochondrial superoxide using cultured neurons. Our findings include that: (i) spinal MT decreased the expression of spinal PGC-1α in the SCDH neurons; (ii) rPGC-1α increased mechanical threshold and thermal latency in MT animals; (iii) Mito-Tempol reduced MT behavioral response; (iv) rPGC-1α reduced MT-induced mitochondria-targeted superoxide; and (v) cultured neuronal cells treated with TNFα increased mitochondria-targeted superoxide that can be inhibited by rPGC-1α. The present findings suggest that spinal PGC-1α reduce MT through decreasing mitochondria-targeted superoxide in the SCDH.
Collapse
Affiliation(s)
- Yuta Kashiwagi
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Hyun Yi
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Keiya Takahashi
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Kentaro Hayashi
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Daigo Ikegami
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Xun Zhu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Jun Gu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, United States.
| |
Collapse
|
9
|
Emery MA, Eitan S. Drug-specific differences in the ability of opioids to manage burn pain. Burns 2019; 46:503-513. [PMID: 31859093 DOI: 10.1016/j.burns.2019.03.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/04/2019] [Accepted: 03/30/2019] [Indexed: 12/17/2022]
Abstract
Burn injury pain is a significant public health problem. Burn injury treatment has improved tremendously in recent decades. However, an unintended consequence is that a larger number of patients now survive more severe injuries, and face intense pain that is very hard to treat. Although many efforts have been made to find alternative treatments, opioids remain the most effective medication available. Burn patients are frequently prescribed opioids in doses and durations that are significantly higher and longer than standard analgesic dosing guidelines. Despite this, many continue to experience unrelieved pain. They are also placed at a higher risk for developing dependence and opioid use disorder. Burn injury profoundly alters the functional state of the immune system. It also alters the expression levels of receptor, effector, and signaling molecules within the spinal cord's dorsal horn. These alterations could explain the reduced potency of opioids. However, recent studies demonstrate that different opioids signal preferentially via differential signaling pathways. This ligand-specific signaling by different opioids implies that burn injury may reduce the antinociceptive potency of opioids to different degrees, in a drug-specific manner. Indeed, recent findings hint at drug-specific differences in the ability of opioids to manage burn pain early after injury, as well as differences in their ability to prevent or treat the development of chronic and neuropathic pain. Here we review the current state of opioid treatment, as well as new findings that could potentially lead to opioid-based pain management strategies that may be significantly more effective than the current solutions.
Collapse
Affiliation(s)
- Michael A Emery
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), USA.
| |
Collapse
|
10
|
Weng Y, Wu J, Li L, Shao J, Li Z, Deng M, Zou W. Circular RNA expression profile in the spinal cord of morphine tolerated rats and screen of putative key circRNAs. Mol Brain 2019; 12:79. [PMID: 31533844 PMCID: PMC6751888 DOI: 10.1186/s13041-019-0498-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/05/2019] [Indexed: 12/17/2022] Open
Abstract
Morphine tolerance developed after repeated or continuous morphine treatment is a global health concern hindering the control of chronic pain. In our previous research, we have reported that the expression of lncRNAs and microRNAs have been greatly modified in the spinal cord of morphine tolerated rats, and the modulating role of miR-873a-5p, miR-219-5p and miR-365 have already been confirmed. However, whether circular RNAs, another essential kind of non-coding RNA, are involved in the pathogenesis of morphine tolerance is still beyond our knowledge. In this study, we conducted microarray analysis for circRNA profile and found a large number of circRNAs changed greatly in the spinal cord by morphine treatment. Among them, we selected nine circRNAs for validation, and seven circRNAs are confirmed. Gene Ontology/Kyoto Encyclopedia of Genes and Genomes (GO/KEGG) analysis were used for functional annotation. Besides, we confirmed the modified expression of seven circRNAs after validation by real-time PCR, selected 3 most prominently modulated ones among them and predicted their downstream miRNA-mRNA network and analyzed their putative function via circRNA-miRNA-mRNA pathway. Finally, we enrolled the differentially expressed mRNAs derived from the identical spinal cord, these validated circRNAs and their putative miRNA targets for ceRNA analysis and screened a promising circRNA-miRNA-mRNA pathway in the development of morphine tolerance. This study, for the first time, provided valuable information on circRNA profile and gave clues for further study on the circRNA mechanism of morphine tolerance.
Collapse
Affiliation(s)
- Yingqi Weng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jing Wu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lin Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Anesthesiology, The First Hospital of Changsha, Changsha, 410008, Hunan, China
| | - Jiali Shao
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Anesthesiology, Hunan Cancer Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhengyiqi Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Meiling Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
11
|
Huang J, Wang J, Guo Q, Zou W. Emerging roles of microRNAs in morphine tolerance. J Pain Res 2019; 12:1139-1147. [PMID: 31114297 PMCID: PMC6497837 DOI: 10.2147/jpr.s187592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/25/2019] [Indexed: 12/19/2022] Open
Abstract
Morphine is commonly used in clinical management to alleviate moderate-to-severe pain. However, prolonged and repeated use of morphine leads to tolerance. Morphine tolerance is a challenging clinical problem that limits its clinical application in pain treatment. The mechanisms underlying morphine tolerance are still not completely understood. MicroRNAs (miRNAs) are small noncoding RNAs containing 18~22 nucleotides that modulate gene expression in a post-transcriptional manner, and their dysregulation causes various diseases. miRNAs bind to the 3ʹ-UTR (untranslated region) of target gene mRNA, inhibiting or destabilizing translation of the transcripts. Morphine causes differential miRNA upregulation or downregulation. This review will present evidence for the contribution of miRNAs to tolerance of the antinociception effect of opioids.
Collapse
Affiliation(s)
- Jiangju Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| |
Collapse
|
12
|
Rashki A, Mumtaz F, Jazayeri F, Shadboorestan A, Esmaeili J, Ejtemaei Mehr S, Ghahremani MH, Dehpour AR. Cyclosporin A attenuating morphine tolerance through inhibiting NO/ERK signaling pathway in human glioblastoma cell line: the involvement of calcineurin. EXCLI JOURNAL 2018; 17:1137-1151. [PMID: 30713473 PMCID: PMC6341459 DOI: 10.17179/excli2018-1693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/06/2018] [Indexed: 12/16/2022]
Abstract
Cyclosporin A (CsA) is known to have an immunosuppressive action. However, it is also attracting attention due to its effects on the nervous system, such as inhibiting the development and expression of morphine-induced tolerance and dependence through unknown mechanisms. It has been shown that CsA modulates the nitric oxide (NO) synthesis and extracellular signal-regulated kinases (ERK) activation, which are potentially involved in signaling pathways in morphine-induced tolerance in cellular models. Therefore, the current study was designed to evaluate the modulatory role of CsA on the MOR tolerance, by targeting the downstream signaling pathway of NO and ERK using an in vitro model. For this purpose, T98G cells were pretreated with CsA, calcineurin autoinhibitory peptide (CAIP), and NG-nitro-l-arginine methyl ester (L-NAME) 30 min before 18 h exposure to MOR. Then, we analyzed the intracellular cyclic adenosine monophosphate (cAMP) levels and also the expression of phosphorylated ERK and nitric oxide synthase (nNOS) proteins. Our results showed that CsA (1 nM, 10 nM, and 100 nM) and CAIP (50 µM) have significantly reduced cAMP and nitrite levels as compared to MOR-treated (2.5 µM) T98G cells. This clearly revealed the attenuation of MOR tolerance by CsA. The expression of nNOS and p-ERK proteins were down-regulated when the T98G cells were pretreated with CsA (1 nM, 10 nM, and 100 nM), CAIP (50 µM), and L-NAME (0.1 mM) as compared to MOR. In conclusion, the CsA pretreatment had a modulatory role in MOR-induced tolerance, which was possibly mediated through NO/ERK signaling pathway.
Collapse
Affiliation(s)
- Asma Rashki
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faiza Mumtaz
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farahnaz Jazayeri
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamileh Esmaeili
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Ejtemaei Mehr
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Liu S, Yao JL, Wan XX, Song ZJ, Miao S, Zhao Y, Wang XL, Liu YP. Sonic hedgehog signaling in spinal cord contributes to morphine-induced hyperalgesia and tolerance through upregulating brain-derived neurotrophic factor expression. J Pain Res 2018; 11:649-659. [PMID: 29662325 PMCID: PMC5892616 DOI: 10.2147/jpr.s153544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Purpose Preventing opioid-induced hyperalgesia and tolerance continues to be a major clinical challenge, and the underlying mechanisms of hyperalgesia and tolerance remain elusive. Here, we investigated the role of sonic hedgehog (Shh) signaling in opioid-induced hyperalgesia and tolerance. Methods Shh signaling expression, behavioral changes, and neurochemical alterations induced by morphine were analyzed in male adult CD-1 mice with repeated administration of morphine. To investigate the contribution of Shh to morphine-induced hyperalgesia (MIH) and tolerance, Shh signaling inhibitor cyclopamine and Shh small interfering RNA (siRNA) were used. To explore the mechanisms of Shh signaling in MIH and tolerance, brain-derived neurotrophic factor (BDNF) inhibitor K252 and anti-BDNF antibody were used. Results Repeated administration of morphine produced obvious hyperalgesia and tolerance. The behavioral changes were correlated with the upregulation and activation of morphine treatment-induced Shh signaling. Pharmacologic and genetic inhibition of Shh signaling significantly delayed the generation of MIH and tolerance and associated neurochemical changes. Chronic morphine administration also induced upregulation of BDNF. Inhibiting BDNF effectively delayed the generation of MIH and tolerance. The upregulation of BDNF induced by morphine was significantly suppressed by inhibiting Shh signaling. In naïve mice, exogenous activation of Shh signaling caused a rapid increase of BDNF expression, as well as thermal hyperalgesia. Inhibiting BDNF significantly suppressed smoothened agonist-induced hyperalgesia. Conclusion These findings suggest that Shh signaling may be a critical mediator for MIH and tolerance by regulating BDNF expression. Inhibiting Shh signaling, especially during the early phase, may effectively delay or suppress MIH and tolerance.
Collapse
Affiliation(s)
- Su Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jun-Li Yao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Anesthesiology, Xuzhou Children's Hospital, Xuzhou, Jiangsu, China
| | - Xin-Xin Wan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhi-Jing Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shuai Miao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ye Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiu-Li Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yue-Peng Liu
- Center of Clinical Research and Translational Medicine, Lianyungang Oriental Hospital, Lianyungang, Jiangsu, China
| |
Collapse
|
14
|
Wang W, Peng Y, Yang H, Bu H, Guo G, Liu D, Shu B, Tian X, Luo A, Zhang X, Gao F. Potential role of CXCL10/CXCR3 signaling in the development of morphine tolerance in periaqueductal gray. Neuropeptides 2017; 65:120-127. [PMID: 28755808 DOI: 10.1016/j.npep.2017.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/16/2017] [Accepted: 07/23/2017] [Indexed: 11/28/2022]
Abstract
Tolerance to morphine antinociception hinders its long-term use in clinical practice. Interaction between neuron and microglia has been proved to play critical role in the mechanism of morphine tolerance, while CXCL10/CXCR3 signaling has been implicated in neuron-glia signaling and morphine analgesia. This study aims to investigate whether CXCL10/CXCR3 signaling in periaqueductal gray (PAG) contributes to the development of morphine tolerance by modulating neuron-microglia interaction. The results showed that the expressions of CXCR3 and CXCL10 were gradually increased in parallel with repeated morphine administration and activation of microglia. CXCR3 was co-localized with neuronal marker NeuN, while CXCL10 was derived from microglia. Microglia inhibitor minocycline significantly attenuated the expression of CXCL10, besides, both minocycline and CXCR3 inhibitor alleviated the development of morphine tolerance. Taken together, our study provided the evidence that CXCL10/CXCR3 signaling in PAG is involved in the development of morphine analgesic tolerance via neuron-microglia interaction.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Yawen Peng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Hui Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Huilian Bu
- Department of Anesthesiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Genhua Guo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Daiqiang Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Bin Shu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China
| | - Xuming Zhang
- School of Life & Health Sciences, Aston University, Aston triangle, Birmingham B4 7ET, United Kingdom
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
15
|
Xiong W, Huang L, Shen Y, Guan S, He L, Tong Z, Tan M, Liu L, Gao Y. Effects of lncRNA uc.48+ siRNA on the release of CGRP in the spinal cords of rats with diabetic neuropathic pain. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:9960-9969. [PMID: 31966886 PMCID: PMC6965966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/24/2017] [Indexed: 06/10/2023]
Abstract
Long noncoding RNA (lncRNA) and factors influencing lncRNA expression are related to the nervous system diseases. The aims of the project are to study the effect of lncRNA uc.48+ siRNA on calcitonin gene related peptide (CGRP) release in the spinal cords (SCs) of diabetic neuropathic pain (DNP) rats to identify its possible mechanism and to provide new experimental evidence for the prevention and treatment of DNP. Male Sprague-Dawley rats were used to create a DNP rat model by feeding the rats a high-fat and fructose diet in addition to an intraperitoneal injection of streptozocin. Fasting blood glucose (FBG), mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) were measured to select the DNP rats. The DNP rats were randomly divided into the following 4 groups: (1) a normal control group (Control), (2) a DNP rats treated with saline group (DNP), (3) a DNP rats treated with uc.48+ siRNA group (DNP + uc.48+ siRNA) and (4) a DNP rats treated with scrambled siRNA group (DNP + scramble siRNA). After intrathecal injection of uc.48+ small interfering RNA, the MWT and TWL of the DNP group significantly decreased compared to the Control group, but after the injection of uc.48+ small interfering RNA, the MWT and TWL of the DNP rats significantly increased (P<0.01, ANOVA test). The application of the methods of qPCR and WB produced results that revealed that the expressions of lncRNA uc.48+, CGRP, IL-1β and TNF-α in the SCs of the DNP group were much higher than those in the Control group (P<0.01, ANOVA test), but the expressions of these molecules in the DNP + uc.48+ siRNA group significantly decreased compared with the DNP group (P<0.01, ANOVA test). The phosphorylations of p38 and ERK1/2 in the DNP group were significantly enhanced compared with the Control group, whereas uc.48+ siRNA significantly reduced the increased phosphorylations of p38 and ERK1/2 pathway in the SCs of the DNP rats (P<0.01, ANOVA test). ELISA results revealed that uc.48+ siRNA significantly decreased the high levels of IL-1β and TNF-α in the sera of the DNP rats (P<0.01, ANOVA test). Therefore, lncRNA uc.48+ may play an important role in the transmission of DNP by promoting the release of CGRP in the SC. Small interfering lncRNA uc.48+ might alleviate the hyperalgesia and allodynia of DNP rats by suppressing the release of CGRP in the SCs of DNP rats, which might inhibit the phosphorylations of p38 and ERK1/2 and suppress the release of IL-1β and TNF-α in the SCs of DNP rats.
Collapse
Affiliation(s)
- Wei Xiong
- Affiliated Stomatological Hospital of Nanchang UniversityNanchang, Jiangxi, P. R. China
- Jiangxi Provincial Key Laboratory of Oral BiomedicineNanchang, Jiangxi, P. R. China
| | - Liping Huang
- Department of Physiology, Medical College of Nanchang UniversityNanchang, Jiangxi, P. R. China
| | - Yulin Shen
- Department of Physiology, Medical College of Nanchang UniversityNanchang, Jiangxi, P. R. China
| | - Shu Guan
- Department of Physiology, Medical College of Nanchang UniversityNanchang, Jiangxi, P. R. China
| | - Lingkun He
- Affiliated Stomatological Hospital of Nanchang UniversityNanchang, Jiangxi, P. R. China
| | - Zhoujie Tong
- Queen Mary College of Grade 2013, Nanchang UniversityNanchang, Jiangxi, P. R. China
| | - Mengxia Tan
- Department of Physiology, Medical College of Nanchang UniversityNanchang, Jiangxi, P. R. China
| | - Lijuan Liu
- Affiliated Stomatological Hospital of Nanchang UniversityNanchang, Jiangxi, P. R. China
| | - Yun Gao
- Department of Physiology, Medical College of Nanchang UniversityNanchang, Jiangxi, P. R. China
- Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and DiseaseNanchang, Jiangxi, P. R. China
| |
Collapse
|
16
|
Walker CS, Raddant AC, Woolley MJ, Russo AF, Hay DL. CGRP receptor antagonist activity of olcegepant depends on the signalling pathway measured. Cephalalgia 2017; 38:437-451. [PMID: 28165287 DOI: 10.1177/0333102417691762] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Calcitonin gene-related peptide (CGRP) is a neuropeptide that acts in the trigeminovascular system and is believed to play an important role in migraine. CGRP activates two receptors that are both present in the trigeminovascular system; the CGRP receptor and the amylin 1 (AMY1) receptor. CGRP receptor antagonists, including olcegepant (BIBN4096BS) and telcagepant (MK-0974), can treat migraine. This study aimed to determine the effectiveness of these antagonists at blocking CGRP receptor signalling in trigeminal ganglia (TG) neurons and transfected CGRP and AMY1 receptors in Cos7 cells, to better understand their mechanism of action. Methods CGRP stimulation of four intracellular signalling molecules relevant to pain (cAMP, CREB, p38 and ERK) were examined in rat TG neurons and compared to transfected CGRP and AMY1 receptors in Cos7 cells. Results In TG neurons, olcegepant displayed signal-specific differences in antagonism of CGRP responses. This effect was also evident in transfected Cos7 cells, where olcegepant blocked CREB phosphorylation more potently than expected at the AMY1 receptor, suggesting that the affinity of this antagonist can be dependent on the signalling pathway activated. Conclusions CGRP receptor antagonist activity appears to be assay-dependent. Thus, these molecules may not be as selective for the CGRP receptor as commonly reported.
Collapse
Affiliation(s)
- Christopher S Walker
- 1 School of Biological Sciences, University of Auckland, Auckland, New Zealand.,2 Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Ann C Raddant
- 3 Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Michael J Woolley
- 4 Institute of Clinical Studies, University of Birmingham, Edgbaston, Birmingham, UK
| | - Andrew F Russo
- 3 Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.,5 Department of Neurology, University of Iowa; Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Debbie L Hay
- 1 School of Biological Sciences, University of Auckland, Auckland, New Zealand.,2 Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
17
|
Yang ZZ, Li L, Wang L, Xu MC, An S, Jiang C, Gu JK, Wang ZJJ, Yu LS, Zeng S. siRNA capsulated brain-targeted nanoparticles specifically knock down OATP2B1 in mice: a mechanism for acute morphine tolerance suppression. Sci Rep 2016; 6:33338. [PMID: 27629937 PMCID: PMC5024137 DOI: 10.1038/srep33338] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/24/2016] [Indexed: 12/18/2022] Open
Abstract
Regulating main brain-uptake transporter of morphine may restrict its tolerance generation, then modify its antinociception. In this study, more than 2 fold higher intracellular uptake concentrations for morphine and morphine-6-glucuronide (M6G) were observed in stable expression cells, HEK293-hOATP2B1 than HEK293-MOCK. Specifically, the Km value of morphine to OATP2B1 (57.58 ± 8.90 μM) is 1.4-time more than that of M6G (80.31 ± 21.75 μM); Cyclosporine A (CsA), an inhibitor of OATP2B1, can inhibit their intracellular accumulations with IC50 = 3.90 ± 0.50 μM for morphine and IC50 = 6.04 ± 0.86 μM for M6G, respectively. To further investigate the role of OATP2B1 in morphine brain transport and tolerance, the novel nanoparticles of DGL-PEG/dermorphin capsulated siRNA (OATP2B1) were applied to deliver siRNA into mouse brain. Along with OATP2B1 depressed, a main reduction was found for each of morphine or M6G in cerebrums or epencephalons of acute morphine tolerance mice. Furthermore, calcium/calmodulin-dependent protein kinase IIα (CaMKIIα) in mouse prefrontal cortex (mPFC) underwent dephosphorylation at Thr286. In conclusion, OATP2B1 downregulation in mouse brain can suppress tolerance via blocking morphine and M6G brain transport. These findings might help to improve the pharmacological effects of morphine.
Collapse
Affiliation(s)
- Zi-Zhao Yang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Li Li
- Zhejiang Provincial Key Laboratory of Geriatrics &Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, 12 Lingyin Road, Hangzhou, Zhejiang Province 310013, China
| | - Lu Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ming-Cheng Xu
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Sai An
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Jing-Kai Gu
- School of Life Sciences, Jilin Univeristy, Changchun, 130012, China
| | - Zai-Jie Jim Wang
- Department of Biopharmaceutical Sciences, University of Illinois, 833 S. Wood Street, Chicago, IL 60612, USA
| | - Lu-Shan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
18
|
Hu XM, Cao SB, Zhang HL, Lyu DM, Chen LP, Xu H, Pan ZQ, Shen W. Downregulation of miR-219 enhances brain-derived neurotrophic factor production in mouse dorsal root ganglia to mediate morphine analgesic tolerance by upregulating CaMKIIγ. Mol Pain 2016; 12:12/0/1744806916666283. [PMID: 27599867 PMCID: PMC5014090 DOI: 10.1177/1744806916666283] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/26/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increasing evidence suggests that microRNAs are functionally involved in the initiation and maintenance of pain hypersensitivity, including chronic morphine analgesic tolerance, through the posttranscriptional regulation of pain-related genes. We have previously demonstrated that miR-219 regulates inflammatory pain in the spinal cord by targeting calcium/calmodulin-dependent protein kinase II gamma (CaMKIIγ). However, whether miR-219 regulates CaMKIIγ expression in the dorsal root ganglia to mediate morphine tolerance remains unclear. RESULTS MiR-219 expression was downregulated and CaMKIIγ expression was upregulated in mouse dorsal root ganglia following chronic morphine treatment. The changes in miR-219 and CaMKIIγ expression closely correlated with the development of morphine tolerance, which was measured using the reduction of percentage of maximum potential efficiency to thermal stimuli. Morphine tolerance was markedly delayed by upregulating miR-219 expression using miR-219 mimics or downregulating CaMKIIγ expression using CaMKIIγ small interfering RNA. The protein and mRNA expression of brain-derived neurotrophic factor were also induced in dorsal root ganglia by prolonged morphine exposure in a time-dependent manner, which were transcriptionally regulated by miR-219 and CaMKIIγ. Scavenging brain-derived neurotrophic factor via tyrosine receptor kinase B-Fc partially attenuated morphine tolerance. Moreover, functional inhibition of miR-219 via miR-219-sponge in naive mice elicited thermal hyperalgesia and spinal neuronal sensitization, which were both suppressed by CaMKIIγ small interfering RNA or tyrosine receptor kinase B-Fc. CONCLUSIONS These results demonstrate that miR-219 contributes to the development of chronic tolerance to morphine analgesia in mouse dorsal root ganglia by targeting CaMKIIγ and enhancing CaMKIIγ-dependent brain-derived neurotrophic factor expression.
Collapse
Affiliation(s)
- Xue-Ming Hu
- Department of Pain Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Shanghai, China Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Shou-Bin Cao
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China Department of Anesthesiology, Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Hai-Long Zhang
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Dong-Mei Lyu
- Department of Pharmacology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Li-Ping Chen
- Department of Pain Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Heng Xu
- Department of Pain Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Zhi-Qiang Pan
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Wen Shen
- Department of Pain Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Ahmadi S, Rashidi A. Gene Expression Profile of Calcium/Calmodulin-Dependent Protein Kinase IIα in Rat Spinal Cord and Midbrain During Induction of Morphine Analgesic Tolerance. ACTA ACUST UNITED AC 2016. [DOI: 10.17795/gct-38142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Zhang H, Li L, Sun Y, Zhang X, Zhang Y, Xu S, Zhao P, Liu T. Sevoflurane prevents stroke-induced depressive and anxiety behaviors by promoting cannabinoid receptor subtype I-dependent interaction between β-arrestin 2 and extracellular signal-regulated kinases 1/2 in the rat hippocampus. J Neurochem 2016; 137:618-29. [PMID: 26991409 DOI: 10.1111/jnc.13613] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/03/2016] [Accepted: 03/09/2016] [Indexed: 01/19/2023]
Abstract
One of the most frequent psychological consequences of stroke is depression. Previous animal studies have demonstrated that post-conditioning with sevoflurane protects against focal cerebral ischemia and reperfusion injury. Thus, we hypothesized that repeated exposure to sevoflurane after transient ischemia can prevent the development of depressive-like behavior. To test this hypothesis, we induced transient cerebral ischemia via transient occlusion of bilateral common carotid arteries and examined the effects of subsequent repeated exposure to sevoflurane on sucrose preference, locomotor activity, and rearing activity in rats. To explore the putative neurobiological mechanisms, we further investigated the roles of hippocampal CB1 receptor in the behavioral effects of sevoflurane. We found that repeated sevoflurane exposures reversed ischemia-induced depressive-like behaviors. Furthermore, CB1 receptor inhibition in the dorsal hippocampus (DH) abolished the effects of sevoflurane exposures on ischemia-induced depressive-like behaviors. In addition, repeated sevoflurane exposures increased CB1 receptor expression and endocannabinoids levels in the DH of ischemic rats. Moreover, repeated sevoflurane exposures enhanced the expression of β-arrestin 2, increased the activation of extracellular signal-regulated kinases (ERK)1/2, and promoted the interaction of β-arrestin 2 and ERK1/2 in the DH, and such effects were reversed by CB1 receptor antagonism in the DH. Finally, β-arrestin 2 expression and ERK1/2 activation in the DH were critical for the preventative effects of sevoflurane exposures on ischemia-induced depressive-like behaviors. Taken together, our results suggested that sevoflurane exposure after brain ischemia may prevent the development of depression, and such preventative effects of sevoflurane are likely ascribed to the activation of CB1 receptor-mediated β-arrestin 2-ERK1/2 signaling pathways. We propose that the following mechanisms are critical for the preventative effects of sevoflurane against post-stroke depressive and anxiety behaviors: repeated sevoflurane exposure after transient brain ischemia enhances N-arachidonoylethanolamine (AEA) and 2-Arachidonoylglycerol (2-AG) levels and normalize cannabinoid receptor type 1 (CB1) receptor expression in the dorsal hippocampus, which results in enhanced interaction of β-arrestin 2 and extracellular signal-regulated kinases (ERK1/2) and increased ERK1/2 activation, leading to decreased depressive and anxiety behaviors. We think these findings should provide a new strategy for treatment of post-stroke depression.
Collapse
Affiliation(s)
- Houzhong Zhang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Li Li
- Department of Anesthesiology, The Obstetrics and Gynecology Hospital of Changchun, Changchun, China
| | - Yanli Sun
- Department of Anesthesiology, The People's Hospital of Changchun City, Changchun, China
| | - Xingyi Zhang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yifan Zhang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Shuang Xu
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Peng Zhao
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Tiecheng Liu
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Guedon JMG, Wu S, Zheng X, Churchill CC, Glorioso JC, Liu CH, Liu S, Vulchanova L, Bekker A, Tao YX, Kinchington PR, Goins WF, Fairbanks CA, Hao S. Current gene therapy using viral vectors for chronic pain. Mol Pain 2015; 11:27. [PMID: 25962909 PMCID: PMC4446851 DOI: 10.1186/s12990-015-0018-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/01/2015] [Indexed: 02/07/2023] Open
Abstract
The complexity of chronic pain and the challenges of pharmacotherapy highlight the importance of development of new approaches to pain management. Gene therapy approaches may be complementary to pharmacotherapy for several advantages. Gene therapy strategies may target specific chronic pain mechanisms in a tissue-specific manner. The present collection of articles features distinct gene therapy approaches targeting specific mechanisms identified as important in the specific pain conditions. Dr. Fairbanks group describes commonly used gene therapeutics (herpes simplex viral vector (HSV) and adeno-associated viral vector (AAV)), and addresses biodistribution and potential neurotoxicity in pre-clinical models of vector delivery. Dr. Tao group addresses that downregulation of a voltage-gated potassium channel (Kv1.2) contributes to the maintenance of neuropathic pain. Alleviation of chronic pain through restoring Kv1.2 expression in sensory neurons is presented in this review. Drs Goins and Kinchington group describes a strategy to use the replication defective HSV vector to deliver two different gene products (enkephalin and TNF soluble receptor) for the treatment of post-herpetic neuralgia. Dr. Hao group addresses the observation that the pro-inflammatory cytokines are an important shared mechanism underlying both neuropathic pain and the development of opioid analgesic tolerance and withdrawal. The use of gene therapy strategies to enhance expression of the anti-pro-inflammatory cytokines is summarized. Development of multiple gene therapy strategies may have the benefit of targeting specific pathologies associated with distinct chronic pain conditions (by Guest Editors, Drs. C. Fairbanks and S. Hao).
Collapse
Affiliation(s)
- Jean-Marc G Guedon
- Graduate Program in Molecular Virology and Microbiology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA. .,Department of Ophthalmology, University of Pittsburgh School of Medicine, Room 1020 EEI, 203 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA.
| | - Xuexing Zheng
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | | | - Joseph C Glorioso
- Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 424 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Ching-Hang Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA.
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA. .,Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA. .,Department of Neurology & Neuroscience, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA. .,Department of Physiology & Pharmacology, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA.
| | - Paul R Kinchington
- Graduate Program in Molecular Virology and Microbiology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA. .,Department of Ophthalmology, University of Pittsburgh School of Medicine, Room 1020 EEI, 203 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - William F Goins
- Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 424 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Carolyn A Fairbanks
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA. .,Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA. .,Department of Pharmacology, University of Minnesota, 9-177 Weaver Densford Hall, 308 Harvard Street, Minneapolis, MN, 55455, USA.
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
22
|
Possible interaction of hippocampal nitric oxide and calcium/calmodulin-dependent protein kinase II on reversal of spatial memory impairment induced by morphine. Eur J Pharmacol 2015; 751:99-111. [DOI: 10.1016/j.ejphar.2015.01.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 01/24/2023]
|
23
|
Dong Y, Li P, Ni Y, Zhao J, Liu Z. Decreased microRNA-125a-3p contributes to upregulation of p38 MAPK in rat trigeminal ganglions with orofacial inflammatory pain. PLoS One 2014; 9:e111594. [PMID: 25380251 PMCID: PMC4224409 DOI: 10.1371/journal.pone.0111594] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 10/03/2014] [Indexed: 11/23/2022] Open
Abstract
Orofacial inflammatory pain is a difficult clinical problem, and the specific molecular mechanisms for this pain remain largely unexplained. The present study aimed to determine the differential expression of microRNAs (miRNAs) and disclose the underlying role of miR-125a-3p in orofacial inflammatory pain induced by complete Freund's adjuvant (CFA). Thirty-two differentially expressed miRNAs were first screened using a microarray chip in ipsilateral trigeminal ganglions (TGs) following CFA injection into the orofacial skin innervated by trigeminal nerve, and a portion of them, including miR-23a*, -24-2*, -26a, -92a, -125a-3p, -183 and -299 were subsequently selected and validated by qPCR. The target genes were predicted based on the miRWalk website and were further analyzed by gene ontology (GO). Further studies revealed miR-125a-3p expression was down-regulated, whereas both the expression of p38 MAPK (mitogen-activated protein kinase) alpha and CGRP (calcitonin gene-related peptide) were up-regulated in ipsilateral TGs at different time points after CFA injection compared with control. Furthermore, mechanistic study revealed that miR-125a-3p negatively regulates p38 alpha gene expression and is positively correlated with the head withdrawal threshold reflecting pain. Luciferase assay showed that binding of miR-125a-3p to the 3′UTR of p38 alpha gene suppressed the transcriptional activity, and overexpression of miR-125a-3p significantly inhibited the p38 alpha mRNA level in ND8/34 cells. Taken together, our results show that miR-125a-3p is negatively correlated with the development and maintenance of orofacial inflammatory pain via regulating p38 MAPK.
Collapse
Affiliation(s)
- Yingchun Dong
- Department of Anesthesiology, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing, China
- * E-mail: (YD); (ZL)
| | - Pengfei Li
- Department of Laboratory, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Yanhong Ni
- Central Laboratory, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing, China
| | - Junjie Zhao
- Department of Periodontics, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing, China
| | - Zhiqiang Liu
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, Houston, United States of America
- * E-mail: (YD); (ZL)
| |
Collapse
|
24
|
Attenuation of Morphine Analgesic Tolerance by Rosuvastatin in Naïve and Morphine Tolerance Rats. Inflammation 2014; 38:134-41. [DOI: 10.1007/s10753-014-0015-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
25
|
Upregulation of pronociceptive mediators and downregulation of opioid peptide by adrenomedullin following chronic exposure to morphine in rats. Neuroscience 2014; 280:31-9. [PMID: 25218960 DOI: 10.1016/j.neuroscience.2014.08.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/28/2014] [Indexed: 01/16/2023]
Abstract
Adrenomedullin (AM) belongs to a calcitonin gene-related peptide (CGRP) family and has been demonstrated to recruit CGRP following chronic use of morphine and neuronal nitric oxide synthase (nNOS) in inflammation. The present study investigated the possibility that AM initiates the changes of other molecules contributing to the development of morphine tolerance in its chronic use. Intrathecal (i.t.) co-administration of the AM receptor antagonist AM22-52 (35.8 μg) inhibited tolerance to morphine-induced analgesia while a daily injection of the AM receptor agonist AM1-50 (8 μg, i.t., bolus) for 9 days induced a decrease in the potency of morphine analgesia and thermal hyperalgesia. Persistent exposure of cultured dorsal root ganglion (DRG) explants to morphine (3.3 μM) for 4 days resulted in an increase in AM and CGRP mRNA levels. However, morphine failed to produce these effects in the presence of AM22-52 (2 μM). The i.t. administration of morphine for 6 days increased the expression of nNOS in the spinal dorsal horn and DRG neurons but decreased expression of the endogenous opioid peptide bovine adrenal medulla 22 (BAM22) in small- and medium-sized neurons in DRG. Particularly, the co-administration of AM22-52 (35.8 μg) inhibited the morphine-induced alterations in nNOS and BAM22. These results indicated that the increase in nNOS and CGRP expressions and the decrease in BAM22 were attributed to the increased AM receptor signaling induced by chronic morphine. The present study supports the hypothesis that the enhancement of AM bioactivity triggered upregulation of pronociceptive mediators and downregulation of pain-inhibiting molecule in a cascade contributing to the development of morphine tolerance.
Collapse
|
26
|
Opiate exposure and withdrawal induces a molecular memory switch in the basolateral amygdala between ERK1/2 and CaMKIIα-dependent signaling substrates. J Neurosci 2013; 33:14693-704. [PMID: 24027270 DOI: 10.1523/jneurosci.1226-13.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Opiate reward memories are powerful triggers for compulsive opiate-seeking behaviors. The basolateral amygdala (BLA) is an important structure for the processing of opiate-related associative memories and is functionally linked to the mesolimbic dopamine (DA) pathway. Transmission through intra-BLA DA D1-like and D2-like receptors independently modulates the formation of opiate reward memories as a function of opiate-exposure state. Thus, in the opiate-naive state, intra-BLA D1 transmission is required for opiate-related memory formation. Once opiate dependence and withdrawal has developed, a functional switch to a DA D2-mediated memory mechanism takes place. However, the downstream molecular signaling events that control this functional switch between intra-BLA DA D1 versus D2 receptor transmission are not currently understood. Using an unbiased place conditioning procedure in rats combined with molecular analyses, we report that opiate reward memory acquisition requires intra-BLA ERK1/2 signaling only in the previously opiate-naive state. However, following chronic opiate exposure and withdrawal, intra-BLA reward memory processing switches to a CaMKIIα-dependent memory substrate. Furthermore, the ability of intra-BLA DA D1 or D2 receptor transmission to modulate the motivational salience of opiates similarly operates through a D1-mediated ERK-dependent mechanism in the opiate-naive state, but switches to a D2-mediated CaMKIIα-dependent mechanism in the dependent/withdrawn state. Protein analysis of BLA tissue revealed a downregulation of ERK1/2 phosphorylation and a dramatic reduction in both total and phosphorylated CaMKIIα signaling, specifically in the opiate-dependent/withdrawn state, demonstrating functional control of ERK1/2-dependent versus CaMKIIα-dependent memory mechanisms within the BLA, controlled by opiate-exposure state.
Collapse
|
27
|
Figueroa JD, Cordero K, Serrano-Illan M, Almeyda A, Baldeosingh K, Almaguel FG, De Leon M. Metabolomics uncovers dietary omega-3 fatty acid-derived metabolites implicated in anti-nociceptive responses after experimental spinal cord injury. Neuroscience 2013; 255:1-18. [PMID: 24042033 DOI: 10.1016/j.neuroscience.2013.09.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 01/23/2023]
Abstract
Chronic neuropathic pain is a frequent comorbidity following spinal cord injury (SCI) and often fails to respond to conventional pain management strategies. Preventive administration of docosahexaenoic acid (DHA) or the consumption of a diet rich in omega-3 polyunsaturated fatty acids (O3PUFAs) confers potent prophylaxis against SCI and improves functional recovery. The present study examines whether this novel dietary strategy provides significant antinociceptive benefits in rats experiencing SCI-induced pain. Rats were fed control chow or chow enriched with O3PUFAs for 8weeks before being subjected to sham or cord contusion surgeries, continuing the same diets after surgery for another 8 more weeks. The paw sensitivity to noxious heat was quantified for at least 8weeks post-SCI using the Hargreaves test. We found that SCI rats consuming the preventive O3PUFA-enriched diet exhibited a significant reduction in thermal hyperalgesia compared to those consuming the normal diet. Functional neurometabolomic profiling revealed a distinctive deregulation in the metabolism of endocannabinoids (eCB) and related N-acyl ethanolamines (NAEs) at 8weeks post-SCI. We found that O3PUFAs consumption led to a robust accumulation of novel NAE precursors, including the glycerophospho-containing docosahexaenoyl ethanolamine (DHEA), docosapentaenoyl ethanolamine (DPEA), and eicosapentaenoyl ethanolamine (EPEA). The tissue levels of these metabolites were significantly correlated with the antihyperalgesic phenotype. In addition, rats consuming the O3PUFA-rich diet showed reduced sprouting of nociceptive fibers containing CGRP and dorsal horn neuron p38 mitogen-activated protein kinase (MAPK) expression, well-established biomarkers of pain. The spinal cord levels of inositols were positively correlated with thermal hyperalgesia, supporting their role as biomarkers of chronic neuropathic pain. Notably, the O3PUFA-rich dietary intervention reduced the levels of these metabolites. Collectively, these results demonstrate the prophylactic value of dietary O3PUFA against SCI-mediated chronic pain.
Collapse
Affiliation(s)
- J D Figueroa
- Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA, United States; Department of Basic Sciences, Loma Linda University, Loma Linda, CA, United States; Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, CA, United States
| | | | | | | | | | | | | |
Collapse
|
28
|
Williams JT, Ingram SL, Henderson G, Chavkin C, von Zastrow M, Schulz S, Koch T, Evans CJ, Christie MJ. Regulation of μ-opioid receptors: desensitization, phosphorylation, internalization, and tolerance. Pharmacol Rev 2013; 65:223-54. [PMID: 23321159 DOI: 10.1124/pr.112.005942] [Citation(s) in RCA: 593] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Morphine and related µ-opioid receptor (MOR) agonists remain among the most effective drugs known for acute relief of severe pain. A major problem in treating painful conditions is that tolerance limits the long-term utility of opioid agonists. Considerable effort has been expended on developing an understanding of the molecular and cellular processes that underlie acute MOR signaling, short-term receptor regulation, and the progression of events that lead to tolerance for different MOR agonists. Although great progress has been made in the past decade, many points of contention and controversy cloud the realization of this progress. This review attempts to clarify some confusion by clearly defining terms, such as desensitization and tolerance, and addressing optimal pharmacological analyses for discerning relative importance of these cellular mechanisms. Cellular and molecular mechanisms regulating MOR function by phosphorylation relative to receptor desensitization and endocytosis are comprehensively reviewed, with an emphasis on agonist-biased regulation and areas where knowledge is lacking or controversial. The implications of these mechanisms for understanding the substantial contribution of MOR signaling to opioid tolerance are then considered in detail. While some functional MOR regulatory mechanisms contributing to tolerance are clearly understood, there are large gaps in understanding the molecular processes responsible for loss of MOR function after chronic exposure to opioids. Further elucidation of the cellular mechanisms that are regulated by opioids will be necessary for the successful development of MOR-based approaches to new pain therapeutics that limit the development of tolerance.
Collapse
Affiliation(s)
- John T Williams
- Vollum Institute, Oregon Health Sciences University, Portland, Oregon, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
This paper is the thirty-fourth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2011 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
30
|
Andersen JM, Klykken C, Mørland J. Long-term methadone treatment reduces phosphorylation of CaMKII in rat brain. J Pharm Pharmacol 2012; 64:843-7. [DOI: 10.1111/j.2042-7158.2012.01469.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Abstract
Objectives
To reveal a possible relationship between a previously reported impairment of novelty seeking in rats exposed to methadone and changes in intracellular molecules related to learning and memory.
Methods
Expression of phosphorylated Ca2+-calmodulin kinase II (pCaMKII), extracellular-signal-regulated kinase 2 (pERK2) and cAMP-responsive element binding protein (pCREB), as well as protein kinase A (PKA), was investigated in rat hippocampus one hour, one day and one week after a three-week methadone administration regime. Studies after an equivalent exposure to morphine, and in the frontal pole, were included for comparison.
Key findings
One day after the last methadone injection the hippocampal level of pCaMKII was significantly reduced. This coincides with a previously reported impairment of novelty seeking. At one hour and one week no significant changes were seen. There was no effect on the other proteins. Morphine affected pCaMKII similarly to methadone. Also in the frontal pole the two drugs reduced pCaMKII one day after the last injection.
Conclusion
The impaired novelty seeking previously found in rats administered methadone for three weeks coincides with a reduced level of pCaMKII in the brain. This finding implies that methadone treatment may affect learning and memory processes, and should stimulate further studies in a field with important knowledge gaps.
Collapse
Affiliation(s)
- Jannike M Andersen
- Division of Forensic Medicine and Drug Abuse Research, Norwegian Institute of Public Health, Oslo, Norway
| | - Christine Klykken
- Division of Forensic Medicine and Drug Abuse Research, Norwegian Institute of Public Health, Oslo, Norway
| | - Jørg Mørland
- Division of Forensic Medicine and Drug Abuse Research, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|