1
|
Christensen CW, Weed SE, Brown TE, Hentges ST. Exploring the role of beta-endorphin in activity-based anorexia in mice. Physiol Rep 2025; 13:e70201. [PMID: 39930661 PMCID: PMC11810985 DOI: 10.14814/phy2.70201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/20/2024] [Accepted: 01/06/2025] [Indexed: 02/14/2025] Open
Abstract
Anorexia nervosa (AN) remains one of the most lethal mental health disorders and is poorly understood from a neurobiological perspective. The most widely used animal model of AN is activity-based anorexia (ABA) where scheduled food presentation leads to a spontaneous maladaptive increase in running-wheel activity and rapid weight loss in rodents, recapitulating specific aspects of AN. Research using the ABA paradigm to probe the role of hedonic and homeostatic circuits has indicated that the hypothalamic proopiomelanocortin (POMC) system may play a role in both the increased activity and reduced food intake observed. Previous work has shown that Pomc mRNA and its peptide product beta-endorphin (β-end) are increased during the onset of ABA. β-end is reinforcing and increases locomotor activity, and mice lacking the mu opioid receptor (MOR), the primary target of β-end, display blunted food-anticipatory activity in the ABA paradigm. Thus, the current work was designed to determine if aspects of ABA would be diminished in mice lacking β-end. We did not find any significant differences in wheel-running, food intake, or body weight loss in β-end knockout mice of either sex during ABA compared to wild-type littermates. Therefore, we conclude that the development of ABA does not require β-end.
Collapse
Affiliation(s)
- Connor W. Christensen
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Samantha E. Weed
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Travis E. Brown
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Shane T. Hentges
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| |
Collapse
|
2
|
Shah DP, Sharma PR, Agarwal R, Barik A. A septo-hypothalamic-medullary circuit directs stress-induced analgesia. eLife 2025; 13:RP96724. [PMID: 39831900 PMCID: PMC11745492 DOI: 10.7554/elife.96724] [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] [Indexed: 01/22/2025] Open
Abstract
Stress is a potent modulator of pain. Specifically, acute stress due to physical restraint induces stress-induced analgesia (SIA). However, where and how acute stress and pain pathways interface in the brain are poorly understood. Here, we describe how the dorsal lateral septum (dLS), a forebrain limbic nucleus, facilitates SIA through its downstream targets in the lateral hypothalamic area (LHA) of mice. Taking advantage of transsynaptic viral-genetic, optogenetic, and chemogenetic techniques, we show that the dLS→LHA circuitry is sufficient to drive analgesia and is required for SIA. Furthermore, our results reveal that the dLS→LHA pathway is opioid-dependent and modulates pain through the pro-nociceptive neurons in the rostral ventromedial medulla (RVM). Remarkably, we found that the inhibitory dLS neurons are recruited specifically when the mice struggle to escape under restraint and, in turn, inhibit excitatory LHA neurons. As a result, the RVM neurons downstream of LHA are disengaged, thus suppressing nociception. Together, we delineate a poly-synaptic pathway that can transform escape behavior in mice under restraint to acute stress into analgesia.
Collapse
Affiliation(s)
| | | | - Rachit Agarwal
- Department of Bioengineering, Indian Institute of ScienceBengaluruIndia
| | - Arnab Barik
- Centre for Neuroscience, Indian Institute of ScienceBengaluruIndia
| |
Collapse
|
3
|
Tseng A, Ahmad SM, Hamid A, Lutfy K. The Role of Endogenous Beta-Endorphin and Enkephalins in the Crosstalk Between Ethanol and Morphine. Pharmaceuticals (Basel) 2025; 18:107. [PMID: 39861169 PMCID: PMC11768102 DOI: 10.3390/ph18010107] [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: 11/28/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background: There is clinical concern about the combined use of alcohol and opiates. Several lines of evidence support an interaction between alcohol and the endogenous opioid system. Thus, we hypothesized that ethanol, by causing the release of opioid peptides, may sensitize the system to the action of exogenous opioids such as morphine. Objectives: In this study, using the place conditioning paradigm, a model of reward, we determined whether a morphine challenge would alter the pre-established preference induced by ethanol conditioning in mice, and whether this response was mediated by the mu opioid receptor (MOP). Given that ethanol exposure stimulates the release of opioid peptides, we also assessed the role of beta-endorphin (β-END) and enkephalins (ENKs) in this response. Methods: Mice lacking MOPs, β-END, and/or ENKs, and their respective wild-type controls were tested for preconditioning place preference on day 1. Mice were then conditioned with ethanol (2 g/kg) versus saline on days 2 to 4 and then tested under a drug-free state for postconditioning place preference on day 5. On day 8, mice received a single injection of morphine (5 mg/kg) and were tested for place preference. On the test days, mice were placed in the central chamber and allowed to explore the chambers. The amount of time that mice spent in the drug-paired chamber was recorded. Results: We found that a challenge dose of morphine given on day 8 enhanced the conditioned place preference (CPP) response in mice previously conditioned with ethanol. This response was abolished in MOP-null mice, confirming the role of MOPs in this response. Although this enhanced response was not altered in mice lacking either β-END or ENKs compared to their wild-type littermates/controls, it was completely blunted in mice lacking both β-END and enkephalins. Conclusions: Together, these results suggest that these opioid peptides jointly mediate the crosstalk between the rewarding actions of morphine and ethanol.
Collapse
Affiliation(s)
| | | | | | - Kabirullah Lutfy
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA; (A.T.); (S.M.A.); (A.H.)
| |
Collapse
|
4
|
Kayama T, Tamura A, Xiaoying T, Tsutsui KI, Kitajo K, Sasaki T. Transformer-based classification of visceral pain-related local field potential patterns in the brain. Sci Rep 2024; 14:24372. [PMID: 39420022 PMCID: PMC11487086 DOI: 10.1038/s41598-024-75616-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Neuronal ensemble activity entrained by local field potential (LFP) patterns underlies a variety of brain functions, including emotion, cognition, and pain perception. Recent advances in machine learning approaches may enable more effective methods for analyzing LFP patterns across multiple brain areas than conventional time-frequency analysis. In this study, we tested the performance of two machine learning algorithms, AlexNet and the Transformer models, to classify LFP patterns in eight pain-related brain regions before and during acetic acid-induced visceral pain behaviors. Over short time windows lasting several seconds, applying AlexNet to LFP power datasets, but not to raw time-series LFP traces from multiple brain areas, successfully achieved superior classification performance compared with simple LFP power analysis. Furthermore, applying the Transformer directly to the raw LFP traces achieved significantly superior classification performance than AlexNet when using LFP power datasets. These results demonstrate the utility of the Transformer in the analysis of neurophysiological signals, and pave the way for its future applications in the decoding of more complex neuronal activity patterns.
Collapse
Affiliation(s)
- Tasuku Kayama
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai, 980-8578, Japan
| | - Atsushi Tamura
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai, 980-8578, Japan
| | - Tuo Xiaoying
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai, 980-8578, Japan
| | - Ken-Ichiro Tsutsui
- Laboratory of Systems Neuroscience, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| | - Keiichi Kitajo
- Division of Neural Dynamics, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, 444-8585, Aichi, Japan
- Physiological Sciences Program, Department of Advanced Studies, Graduate University for Advanced Studies (SOKENDAI), 38 Nishigonaka, Myodaiji, Okazaki, 444-8585, Aichi, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai, 980-8578, Japan.
- Department of Neuropharmacology, Tohoku University School of Medicine, 4-1 Seiryo-machi, Aoba-Ku, Sendai, 980-8575, Japan.
| |
Collapse
|
5
|
Rojo D, Hael CE, Soria A, de Souza FSJ, Low MJ, Franchini LF, Rubinstein M. A mammalian tripartite enhancer cluster controls hypothalamic Pomc expression, food intake, and body weight. Proc Natl Acad Sci U S A 2024; 121:e2322692121. [PMID: 38652744 PMCID: PMC11067048 DOI: 10.1073/pnas.2322692121] [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: 01/05/2024] [Accepted: 03/12/2024] [Indexed: 04/25/2024] Open
Abstract
Food intake and energy balance are tightly regulated by a group of hypothalamic arcuate neurons expressing the proopiomelanocortin (POMC) gene. In mammals, arcuate-specific POMC expression is driven by two cis-acting transcriptional enhancers known as nPE1 and nPE2. Because mutant mice lacking these two enhancers still showed hypothalamic Pomc mRNA, we searched for additional elements contributing to arcuate Pomc expression. By combining molecular evolution with reporter gene expression in transgenic zebrafish and mice, here, we identified a mammalian arcuate-specific Pomc enhancer that we named nPE3, carrying several binding sites also present in nPE1 and nPE2 for transcription factors known to activate neuronal Pomc expression, such as ISL1, NKX2.1, and ERα. We found that nPE3 originated in the lineage leading to placental mammals and remained under purifying selection in all mammalian orders, although it was lost in Simiiformes (monkeys, apes, and humans) following a unique segmental deletion event. Interestingly, ablation of nPE3 from the mouse genome led to a drastic reduction (>70%) in hypothalamic Pomc mRNA during development and only moderate (<33%) in adult mice. Comparison between double (nPE1 and nPE2) and triple (nPE1, nPE2, and nPE3) enhancer mutants revealed the relative contribution of nPE3 to hypothalamic Pomc expression and its importance in the control of food intake and adiposity in male and female mice. Altogether, these results demonstrate that nPE3 integrates a tripartite cluster of partially redundant enhancers that originated upon a triple convergent evolutionary process in mammals and that is critical for hypothalamic Pomc expression and body weight homeostasis.
Collapse
Affiliation(s)
- Daniela Rojo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires1428, Argentina
| | - Clara E. Hael
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires1428, Argentina
| | - Agustina Soria
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires1428, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires1428, Argentina
| | - Flávio S. J. de Souza
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires1428, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires and Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires1428, Argentina
| | - Malcolm J. Low
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI48105
| | - Lucía F. Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires1428, Argentina
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires1428, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires1428, Argentina
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI48105
| |
Collapse
|
6
|
Waldron MA, Jones HE, Rhinehart EM, Grisel JE. Sensitivity to the initial rewarding effects of alcohol: Influence of age, sex, and β-endorphin. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:667-679. [PMID: 38426214 DOI: 10.1111/acer.15281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Alcohol use disorders (AUDs) are widespread, devastating and complex. About 20% of people who consume alcohol develop problem use, accounting for over 5% of worldwide deaths. While numerous animal models have facilitated understanding of the consequences of excessive drinking, translational models allow for experimental manipulation of factors thought to contribute to AUD liability. METHODS We employ a single-exposure conditioned place preference assay (SE-CPP) to investigate the influence of age, sex and the opioid peptide β-endorphin (bE) on the initial rewarding effects of ethanol, a strong predictor of AUDs. Adolescent (PND28-35) and adult (PND70-90) male and female, control C57BL/6J and bE-deficient mice were tested following a single injection of 1.5 g/kg of ethanol. Following the SE-CPP test, animals were deeply anesthetized, sacrificed, and perfused, and the brains were subsequently sectioned at 40 microns and processed for immunohistochemical localization of c-fos. One-sample, two-tailed t-tests were used to assess drug preference or aversion and the locomotor effects of alcohol. RESULTS In general, adults were more sensitive to the effects of alcohol than adolescents, and outcomes depended on sex and bE. For example, among females, adolescents were stimulated by the drug, but insensitive to locomotor effects as adults, while among males, adolescents were insensitive and adults sedated. Wild-type adolescents of both sexes failed to evince initial subjective reward from the drug, but bE-deficient adolescents, and all adult subjects, preferred a context once associated with ethanol over one that had been paired with saline. c-fos immunoreactivity in multiple brain regions was attenuated in bE-deficient animals, though influences of both sex and bE grew stronger with age. CONCLUSIONS This study demonstrates the utility of the SE-CPP paradigm for elucidating factors that contribute to the liability for AUDs, and supports the growing body of research that shows that sensitivity to the rewarding effects of alcohol changes during the course of development. Our results also suggest that developmental contributions are sex-dependent, and may also depend on the influence of endogenous opioid signaling.
Collapse
Affiliation(s)
- Madison A Waldron
- Department of Psychology, Neuroscience Program, Bucknell University, Lewisburg, Pennsylvania, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Holly E Jones
- Department of Psychology, Neuroscience Program, Bucknell University, Lewisburg, Pennsylvania, USA
| | - Erin M Rhinehart
- Department of Biology, Susquehanna University, Selinsgrove, Pennsylvania, USA
| | - Judith E Grisel
- Department of Psychology, Neuroscience Program, Bucknell University, Lewisburg, Pennsylvania, USA
| |
Collapse
|
7
|
Stea SG, Grisel JE. β-Endorphin influences sedative and ataxic effects of alcohol. Alcohol 2024; 115:69-77. [PMID: 37741556 DOI: 10.1016/j.alcohol.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Beta-endorphin (β-E) is an opioid peptide linked to the behavioral effects of ethanol. For example, β-E provides negative feedback to inhibit the hypothalamic-pituitary-adrenal (HPA) stress axis, and neuroadaptation of this system to ethanol may facilitate sex differences in disordered drinking. Locomotor sensitivity to ethanol may also influence the risk for addiction; however, the role of β-E in psychomotor effects of ethanol is not fully understood. We examined the role of β-E and sex on locomotor effects of ethanol using adult male and female wild-type C57BL/6J and β-E deficient B6.129S2-Pomctm1Low/J mice in a parallel rod floor apparatus following 0.75 or 2.0 g/kg ethanol. Beginning 15 min after intraperitoneal injection, we recorded foot slips, distance traveled, slips per meter, first instance of immobility, and total time spent off-balance (lying on the floor) over 15 min, and collected blood for analysis of ethanol concentration 60 min after injection. Overall, β-E deficient mice were more sedated and ataxic following ethanol; at the lower dose they slipped more frequently and had a higher rate of slips per meter traveled. At the higher dose, β-E deficient mice were predominantly sedated, slipping less frequently, and traveling less, as well as spending more time off-balance and becoming immobile sooner. Genotype interacted with sex in that male β-E deficient mice slipped more frequently than their female counterparts, suggesting that β-E may elicit sex-dependent effects of ethanol-induced ataxia. Blood ethanol concentration did not differ between any group, suggesting that behavioral differences result from altered sensitivity to ethanol. Our data support the contention that β-E modulates the locomotor effects of ethanol and may influence ataxia in a sex-dependent manner. These findings help elucidate the role of β-E in diverging behavioral responses to ethanol and may aid the development of targeted treatments for alcohol use disorders.
Collapse
Affiliation(s)
- Samuel G Stea
- Department of Psychology & Neuroscience Program, Bucknell University, Lewisburg, PA 17837, United States
| | - Judith E Grisel
- Department of Psychology & Neuroscience Program, Bucknell University, Lewisburg, PA 17837, United States.
| |
Collapse
|
8
|
Ueda H, Neyama H. Fibromyalgia Animal Models Using Intermittent Cold and Psychological Stress. Biomedicines 2023; 12:56. [PMID: 38255163 PMCID: PMC10813244 DOI: 10.3390/biomedicines12010056] [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: 11/14/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Fibromyalgia (FM) is a chronic pain condition characterized by widespread musculoskeletal pain and other frequent symptoms such as fatigue, sleep disturbance, cognitive impairment, and mood disorder. Based on the view that intermittent stress would be the most probable etiology for FM, intermittent cold- and intermittent psychological stress-induced generalized pain (ICGP and IPGP) models in mice have been developed and validated as FM-like pain models in terms of the patho-physiological and pharmacotherapeutic features that are shared with clinical versions. Both models show long-lasting and generalized pain and female-predominant sex differences after gonadectomy. Like many other neuropathic pain models, ICGP and IPGP were abolished in lysophosphatidic acid receptor 1 (LPAR1) knock-out mice or by LPAR1 antagonist treatments, although deciding the clinical importance of this mechanism depends on waiting for the development of a clinically available LPAR1 antagonist. On the other hand, the nonsteroidal anti-inflammatory drug diclofenac with morphine did not suppress hyperalgesia in these models, and this is consistent with the clinical findings. Pharmacological studies suggest that the lack of morphine analgesia is associated with opioid tolerance upon the stress-induced release of endorphins and subsequent counterbalance through anti-opioid NMDA receptor mechanisms. Regarding pharmacotherapy, hyperalgesia in both models was suppressed by pregabalin and duloxetine, which have been approved for FM treatment in clinic. Notably, repeated treatments with mirtazapine, an α2 adrenergic receptor antagonist-type antidepressant, and donepezil, a drug for treating Alzheimer's disease, showed potent therapeutic actions in these models. However, the pharmacotherapeutic treatment should be carried out 3 months after stress, which is stated in the FM guideline, and many preclinical studies, such as those analyzing molecular and cellular mechanisms, as well as additional evidence using different animal models, are required. Thus, the ICGP and IPGP models have the potential to help discover and characterize new therapeutic medicines that might be used for the radical treatment of FM, although there are several limitations to be overcome.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan;
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114201, Taiwan
| | - Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan;
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
9
|
Scopano MR, Jones HE, Stea SG, Freeman MZ, Grisel JE. Age, β-endorphin, and sex dependent effects of maternal separation on locomotor activity, anxiety-like behavior, and alcohol reward. Front Behav Neurosci 2023; 17:1155647. [PMID: 37091593 PMCID: PMC10113444 DOI: 10.3389/fnbeh.2023.1155647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/07/2023] [Indexed: 04/09/2023] Open
Abstract
IntroductionChildhood adversity is pervasive and linked to numerous disadvantages in adulthood, including physical health problems, mental illness, and substance use disorders. Initial sensitivity to the rewarding effects of alcohol predicts the risk of developing an alcohol use disorder, and may be linked to developmental stress. The opioid peptide β-endorphin (β-E) regulates the stress response and is also implicated in the risk for excessive alcohol consumption.MethodsWe explored the influence of β-E in an animal model of early life adversity using controlled maternal separation by evaluating changes in locomotor activity, anxiety-like behavior, and the initial rewarding effects of alcohol in a single exposure conditioned place preference paradigm in control C57BL/6J and β-E deficient β-E +/+ 0.129S2-Pomc tm1Low/J; β-E −/− mice. Maternal separation (MS) occurred for 3 h each day from post-natal days (PND) 5–18 in approximately half the subjects.ResultsMaternal interactions increased following the separation protocol equally in both genotypes. MS and control subjects were tested as adolescents (PND 26–32) or adults (PND 58–72); the effects of MS were generally more pronounced in older subjects. Adults were more active than adolescents in the open field, and MS decreased activity in adolescent mice but increased it in adults. The increase in adult activity as a result of early life stress depended on both β-E and sex. β-E also influenced the effect of maternal separation on anxiety-like behavior in the Elevated Plus Maze. MS promoted rewarding effects of alcohol in male β-E deficient mice of either age, but had no effect in other groups.DiscussionTaken together, these results suggest that the effects of MS develop over time and are β-E and sex dependent and may aid understanding of how individual differences influence the impact of adverse childhood experiences.
Collapse
|
10
|
Lee MT, Mackie K, Chiou LC. Alternative pain management via endocannabinoids in the time of the opioid epidemic: Peripheral neuromodulation and pharmacological interventions. Br J Pharmacol 2023; 180:894-909. [PMID: 34877650 PMCID: PMC9170838 DOI: 10.1111/bph.15771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 01/18/2023] Open
Abstract
The use of opioids in pain management is hampered by the emergence of analgesic tolerance, which leads to increased dosing and side effects, both of which have contributed to the opioid epidemic. One promising potential approach to limit opioid analgesic tolerance is activating the endocannabinoid system in the CNS, via activation of CB1 receptors in the descending pain inhibitory pathway. In this review, we first discuss preclinical and clinical evidence revealing the potential of pharmacological activation of CB1 receptors in modulating opioid tolerance, including activation by phytocannabinoids, synthetic CB1 receptor agonists, endocannabinoid degradation enzyme inhibitors, and recently discovered positive allosteric modulators of CB1 receptors. On the other hand, as non-pharmacological pain relief is advocated by the US-NIH to combat the opioid epidemic, we also discuss contributions of peripheral neuromodulation, involving the electrostimulation of peripheral nerves, in addressing chronic pain and opioid tolerance. The involvement of supraspinal endocannabinoid systems in peripheral neuromodulation-induced analgesia is also discussed. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Grants
- MOST 108-2321-B-002-005 Ministry of Science and Technology, Taiwan
- MOST 107-2811-B-002-008 Ministry of Science and Technology, Taiwan
- R01 DA041229 NIDA NIH HHS
- MOST 107-2321-B-002-010 Ministry of Science and Technology, Taiwan
- R01 DA047858 NIDA NIH HHS
- 107M4022-3 Ministry of Education, Taiwan
- MOST 106-2321-B-002-019 Ministry of Science and Technology, Taiwan
- NHRI-EX111-11114NI National Health Research Institutes, Taiwan
- FRGS/1/2021/WAB13/UCSI/02/1 Ministry of Higher Education, Malaysia
- R21 DA042584 NIDA NIH HHS
- REIG-FPS-2020/065 UCSI University Research Excellence and Innovation Grant, Malaysia
- NHRI-EX109-10733NI National Health Research Institutes, Taiwan
- MOST 104-2745-B-002-004 Ministry of Science and Technology, Taiwan
- MOST 109-2320-B-002-042-MY3 Ministry of Science and Technology, Taiwan
- MOST 107-2811-B-002 -008 Ministry of Science and Technology, Taiwan
- MOST 108-2320-B-002-029-MY3 Ministry of Science and Technology, Taiwan
Collapse
Affiliation(s)
- Ming Tatt Lee
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Ken Mackie
- Gill Center for Biomolecular Research, Indiana University, Bloomington, Indiana 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, USA
| | - Lih-Chu Chiou
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
11
|
Rauchbauer B, Jank G, Dunbar RIM, Lamm C. Only empathy-related traits, not being mimicked or endorphin release, influence social closeness and prosocial behavior. Sci Rep 2023; 13:4072. [PMID: 36906682 PMCID: PMC10008555 DOI: 10.1038/s41598-023-30946-9] [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: 10/26/2022] [Accepted: 03/03/2023] [Indexed: 03/13/2023] Open
Abstract
Seminal studies suggest that being mimicked increases experienced social closeness and prosocial behavior to a mimicking confederate (i.e., interaction partner). Here we reexamine these results by considering the role of empathy-related traits, an indirect proxy for endorphin uptake, and their combined effects as an explanation for these results. 180 female participants were mimicked or anti-mimicked in an interaction with a confederate. The effects of being mimicked versus anti-mimicked in relation to empathy-related traits and endorphin release (assessed indirectly via pain tolerance) on experienced closeness and prosocial behavior were assessed using Bayesian analyses. Our results suggest that high individual empathy-related traits increase social closeness to the anti-mimicking and mimicking confederate and to one's romantic partner, as compared to mimicry alone. Results furthermore strongly suggest that high individual empathy-related traits increase prosocial behavior (donations and willingness to help) as compared to mimicry alone. These findings extend previous work by highlighting that empathy-related traits are more influential in creating positive effects on social closeness and prosocial behavior than a one-shot mimicking encounter.
Collapse
Affiliation(s)
- Birgit Rauchbauer
- Social, Cognitive and Affective Neuroscience Unit, Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, Liebiggasse 5, 1010, Vienna, Austria.,Vienna Cognitive Science Hub, University of Vienna, Universitätsstraße 7, 1010, Vienna, Austria.,Laboratoire Parole et Langage, Aix-Marseille Université, CNRS, 5 Avenue Pasteur, 13100, Aix-en-Provence, France.,Laboratoire de Neurosciences Cognitives, Aix-Marseille Université, CNRS, 3, Place Victor-Hugo, 13331, Marseille Cedex 3, France.,Institute of Language, Communication and the Brain, 13100, Aix-en-Provence, France
| | - Gabriela Jank
- Social, Cognitive and Affective Neuroscience Unit, Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, Liebiggasse 5, 1010, Vienna, Austria
| | - Robin I M Dunbar
- Social and Evolutionary Neuroscience Research Group, Department of Experimental Psychology, University of Oxford, Anna Watts Building, Radcliffe Observatory Quarter, Woodstock Rd, Oxford, OX26GG, UK
| | - Claus Lamm
- Social, Cognitive and Affective Neuroscience Unit, Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, Liebiggasse 5, 1010, Vienna, Austria. .,Vienna Cognitive Science Hub, University of Vienna, Universitätsstraße 7, 1010, Vienna, Austria.
| |
Collapse
|
12
|
Tolentino L, Iqbal A, Rahman S, Lutfy K. The Role of Beta-Endorphin in Food Deprivation-Mediated Increases in Food Intake and Binge-Eating. Brain Sci 2023; 13:brainsci13020212. [PMID: 36831755 PMCID: PMC9954518 DOI: 10.3390/brainsci13020212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Food deprivation and binge eating represent significant public health concerns. Previous studies have implicated that hypothalamic opioids are affected following food deprivation. However, the role of each opioid peptide is not fully understood. Therefore, we investigated the role of endogenous beta-endorphin in food deprivation-mediated increases in food intake and binge eating. Male mice lacking beta-endorphin and their respective controls were subjected to 24 h food deprivation and then were randomly assigned to receive a regular diet (RD) or a high-fat diet (HFD). After four to five weeks, animals were re-exposed to an HFD to assess if previous exposure to HFD would enhance binge-eating behavior. We report that food deprivation significantly increases food intake; however, beta-endorphin may not be involved in this process. In addition, our findings suggest that prior exposure to an HFD promotes binge-eating behavior in wildtype mice, and that these effects were modestly decreased in beta-endorphin knockout mice. Overall, our results support that beta-endorphin may play a modest role in mediating palatability-driven feeding, but not hunger-associated feeding. A better understanding of neural mechanisms involved in binge eating and deprivation-induced increases in food intake may inspire new prevention or treatment options to decrease the burden of eating disorders.
Collapse
Affiliation(s)
- Laica Tolentino
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Asif Iqbal
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, CT 06269, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA
| | - Kabirullah Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA
- Correspondence:
| |
Collapse
|
13
|
Hermann AL, Fell GL, Kemény LV, Fung CY, Held KD, Biggs PJ, Rivera PD, Bilbo SD, Igras V, Willers H, Kung J, Gheorghiu L, Hideghéty K, Mao J, Woolf CJ, Fisher DE. β-Endorphin mediates radiation therapy fatigue. SCIENCE ADVANCES 2022; 8:eabn6025. [PMID: 36525492 PMCID: PMC9757747 DOI: 10.1126/sciadv.abn6025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 11/15/2022] [Indexed: 06/17/2023]
Abstract
Fatigue is a common adverse effect of external beam radiation therapy in cancer patients. Mechanisms causing radiation fatigue remain unclear, although linkage to skin irradiation has been suggested. β-Endorphin, an endogenous opioid, is synthesized in skin following genotoxic ultraviolet irradiation and acts systemically, producing addiction. Exogenous opiates with the same receptor activity as β-endorphin can cause fatigue. Using rodent models of radiation therapy, exposing tails and sparing vital organs, we tested whether skin-derived β-endorphin contributes to radiation-induced fatigue. Over a 6-week radiation regimen, plasma β-endorphin increased in rats, paralleled by opiate phenotypes (elevated pain thresholds, Straub tail) and fatigue-like behavior, which was reversed in animals treated by the opiate antagonist naloxone. Mechanistically, all these phenotypes were blocked by opiate antagonist treatment and were undetected in either β-endorphin knockout mice or mice lacking keratinocyte p53 expression. These findings implicate skin-derived β-endorphin in systemic effects of radiation therapy. Opioid antagonism may warrant testing in humans as treatment or prevention of radiation-induced fatigue.
Collapse
Affiliation(s)
- Andrea L. Hermann
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Oncotherapy, Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Gillian L. Fell
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lajos V. Kemény
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- HCEMM-SU Translational Dermatology Research Group, Department of Physiology, Semmelweis University, Budapest, Hungary
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Claire Y. Fung
- Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Kathryn D. Held
- Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- National Council on Radiation Protection and Measurements, 7910 Woodmont Ave, Suite 400, Bethesda, MD 20814, USA
| | - Peter J. Biggs
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Phillip D. Rivera
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02421, USA
- Department of Biology, Hope College, Holland, MI 49423, USA
| | - Staci D. Bilbo
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA 02421, USA
| | - Vivien Igras
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jong Kung
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Liliana Gheorghiu
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Katalin Hideghéty
- Department of Oncotherapy, Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
- ELI-ALPS Non Profit Ltd., Szeged, Hungary
| | - Jianren Mao
- MGH Center for Translational Pain Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Clifford J. Woolf
- FM Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - David E. Fisher
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
14
|
Patra PH, Tench B, Hitrec T, Holmes F, Drake R, Cerritelli S, Spanswick D, Pickering AE. Pro-Opiomelanocortin (POMC) neurons in the nucleus of the solitary tract mediate endorphinergic endogenous analgesia in mice. Pain 2022; 164:1051-1066. [PMID: 36448978 DOI: 10.1097/j.pain.0000000000002802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 09/27/2022] [Indexed: 12/02/2022]
Abstract
ABSTRACT The nucleus of the solitary tract (NTS) contains pro-opiomelanocortin (POMC) neurons which are one of the two major sources of β-endorphin in the brain. The functional role of these NTS POMC neurons in nociceptive and cardiorespiratory function is debated. We have shown that NTS POMC optogenetic activation produces bradycardia and transient apnoea in a working heart brainstem preparation and chemogenetic activation with an engineered ion channel (PSAM) produced opioidergic analgesia in vivo . To better define the role of the NTS POMC neurons in behaving animals, we adopted in vivo optogenetics (ChrimsonR) and excitatory/inhibitory chemogenetic DREADD (hM3Dq/hM4Di) strategies in POMC-Cre mice. We show that optogenetic activation of NTS POMC neurons produces time-locked, graded, transient bradycardia and bradypnoea in anaesthetised mice which is naloxone sensitive (1 mg/kg, i.p) suggesting a role of β-endorphin. Both optogenetic and chemogenetic activation of NTS POMC neurons produces sustained thermal analgesia in behaving mice which can be blocked by naloxone. It also produced analgesia in inflammatory pain (carrageenan) but not in a neuropathic pain model (tibial nerve transection). Inhibiting NTS POMC neurons does not produce any effect on basal nociception but inhibits stress-induced analgesia (unlike inhibition of arcuate POMC neurons). Activation of NTS POMC neuronal populations in conscious mice did not cause respiratory depression, anxiety or locomotor deficit (in open field) nor affective preference. These findings indicate that NTS POMC neurons play a key role in the generation of endorphinergic endogenous analgesia and can also regulate cardiorespiratory function.
Collapse
Affiliation(s)
- Pabitra Hriday Patra
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Becks Tench
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Timna Hitrec
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Fiona Holmes
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Robert Drake
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Serena Cerritelli
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - David Spanswick
- Neurosolutions, University of Warwick, Gibbet Hill Road, Coventry, West Midlands, CV4 7AL, UK
| | - Anthony Edward Pickering
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| |
Collapse
|
15
|
Yu M, Bean JC, Liu H, He Y, Yang Y, Cai X, Yu K, Pei Z, Liu H, Tu L, Conde KM, Wang M, Li Y, Yin N, Zhang N, Han J, Scarcelli NA, Xu P, He Y, Xu Y, Wang C. SK3 in POMC neurons plays a sexually dimorphic role in energy and glucose homeostasis. Cell Biosci 2022; 12:170. [PMID: 36210455 PMCID: PMC9549684 DOI: 10.1186/s13578-022-00907-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Pro-opiomelanocortin (POMC) neurons play a sexually dimorphic role in body weight and glucose balance. However, the mechanisms for the sex differences in POMC neuron functions are not fully understood. RESULTS We detected small conductance calcium-activated potassium (SK) current in POMC neurons. Secondary analysis of published single-cell RNA-Seq data showed that POMC neurons abundantly express SK3, one SK channel subunit. To test whether SK3 in POMC neurons regulates POMC neuron functions on energy and glucose homeostasis, we used a Cre-loxP strategy to delete SK3 specifically from mature POMC neurons. POMC-specific deletion of SK3 did not affect body weight in either male or female mice. Interestingly, male mutant mice showed not only decreased food intake but also decreased physical activity, resulting in unchanged body weight. Further, POMC-specific SK3 deficiency impaired glucose balance specifically in female mice but not in male mice. Finally, no sex differences were detected in the expression of SK3 and SK current in total POMC neurons. However, we found higher SK current but lower SK3 positive neuron population in male POMC neurons co-expressing estrogen receptor α (ERα) compared to that in females. CONCLUSION These results revealed a sexually dimorphic role of SK3 in POMC neurons in both energy and glucose homeostasis independent of body weight control, which was associated with the sex difference of SK current in a subpopulation of POMC + ERα + neurons.
Collapse
Affiliation(s)
- Meng Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Jonathan C Bean
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Hailan Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yang He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Xing Cai
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kaifan Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Zhou Pei
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Hesong Liu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Longlong Tu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kristine M Conde
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Mengjie Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yongxiang Li
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Na Yin
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Nan Zhang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Junying Han
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Nikolas A Scarcelli
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Pingwen Xu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Brain glycemic and metabolism control department, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
O’Brien C, Vemireddy R, Mohammed U, Barker DJ. Stress reveals a specific behavioral phenotype for opioid abuse susceptibility. J Exp Anal Behav 2022; 117:518-531. [PMID: 35119105 PMCID: PMC9090955 DOI: 10.1002/jeab.738] [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: 10/08/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 11/07/2022]
Abstract
Susceptibility to stress has long been considered important for the development of substance use disorders. Nonetheless, behavioral and physiological responses to stress are highly variable, making it difficult to identify the individuals who are most likely to abuse drugs. In the present study, we employed a comprehensive battery of tests for negative valence behaviors and nociception to identify individuals predisposed to opioid seeking following oral opioid self-administration. Furthermore, we examined how this profile was affected by a history of stress. We observed that mice receiving foot shock stress failed to exhibit a preference for sucrose, showed increased immobility in the forced swim task, and exhibited mechanical hypersensitivity when compared to controls. When considering these behaviors in light of future fentanyl-seeking responses, we observed that heightened mechanical sensitivity corresponded to higher opioid preference in mice with a history of stress, but not controls. Moreover, we were surprised to discover that paradoxically high sucrose preferences predicted fentanyl preference in shock mice, while signs of anhedonia predicted fentanyl preference in controls. Taken together, these results indicate that stress can act as a physiological modulator, shifting profiles of opioid abuse susceptibility depending on an individual's history.
Collapse
Affiliation(s)
- Chris O’Brien
- Department of Psychology, Rutgers, The State University of New Jersey
| | - Roshni Vemireddy
- Department of Psychology, Rutgers, The State University of New Jersey
| | - Uzma Mohammed
- Department of Psychology, Rutgers, The State University of New Jersey
| | - David J. Barker
- Department of Psychology, Rutgers, The State University of New Jersey
- Brain Health Institute, Rutgers University, Piscataway, NJ
| |
Collapse
|
17
|
Weng Y, Yi C, Liang H, Lin K, Zheng X, Xiao J, Han H. The Brain Structural-Functional Vulnerability in Drug-Naive Children With Juvenile Idiopathic Arthritis: Insights From the Hippocampus. Front Hum Neurosci 2022; 16:833602. [PMID: 35370580 PMCID: PMC8973270 DOI: 10.3389/fnhum.2022.833602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/28/2022] [Indexed: 11/26/2022] Open
Abstract
Objective Leveraging an integrative multimodal MRI paradigm to elaborate on the hippocampus-derived structural and functional changes in children and adolescents with juvenile idiopathic arthritis (JIA) and to explore potential correlations within the “joint-inflammation-brain” axis during the period of central neural system (CNS) development. Methods Twenty-one patients with JIA all completed the multimodal MRI scanning, laboratory tests, and neuropsychological assessments; meanwhile, 23 matched controls were recruited. We then harnessed the spherical harmonics with a point distribution model (SPHARM-PDM) and the ROI-to-voxel functional connectivity (FC) to measure the hippocampal shape and hippocampo-cortical FC patterns. Correlation analysis was performed to explore the potential links in neuroimaging features with disease-related indices. Results Compared to controls, JIA patients only presented an atrophic tendency in the posterior part of the bilateral hippocampus. The hippocampo-cortical FC revealed the between-group divergences mainly located at the pain matrix, striatum, and temporal lobe. Remarkably, the enhanced FC between the right hippocampus and postcentral cortex is positively correlated with the disability index, while the weakened FC of right anterior hippocampus with right insula and that of left posterior hippocampus with left superior temporal gyrus was inversely related to the erythrocyte sedimentation rate and anxiety status, separately. Conclusion As with macroscopic damages, the altered functional-connectome patterns of the hippocampus in JIA patients might be more sensitive to detect the early neuropathological changes. Moreover, the functional disturbances were demonstrated associated with the physical disability, inflammation, and emotional status. These findings may enlighten us on the underlying neuropathological mechanism of CNS comorbidities in JIA.
Collapse
Affiliation(s)
- Yifei Weng
- Department of Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Cuili Yi
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hongyan Liang
- Department of Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Kezhao Lin
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaohuang Zheng
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jihong Xiao
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Jihong Xiao,
| | - Haiwei Han
- Department of Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Haiwei Han, ; orcid.org/0000-0003-3608-4931
| |
Collapse
|
18
|
Yu H, Rubinstein M, Low MJ. Developmental single-cell transcriptomics of hypothalamic POMC neurons reveal the genetic trajectories of multiple neuropeptidergic phenotypes. eLife 2022; 11:e72883. [PMID: 35044906 PMCID: PMC8806186 DOI: 10.7554/elife.72883] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 01/18/2022] [Indexed: 11/21/2022] Open
Abstract
Proopiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus are essential to regulate food intake and energy balance. However, the ontogenetic transcriptional programs that specify the identity and functioning of these neurons are poorly understood. Here, we use single-cell RNA-sequencing (scRNA-seq) to define the transcriptomes characterizing Pomc-expressing cells in the developing hypothalamus and translating ribosome affinity purification with RNA-sequencing (TRAP-seq) to analyze the subsequent translatomes of mature POMC neurons. Our data showed that Pomc-expressing neurons give rise to multiple developmental pathways expressing different levels of Pomc and unique combinations of transcription factors. The predominant cluster, featured by high levels of Pomc and Prdm12 transcripts, represents the canonical arcuate POMC neurons. Additional cell clusters expressing medium or low levels of Pomc mature into different neuronal phenotypes featured by distinct sets of transcription factors, neuropeptides, processing enzymes, cell surface, and nuclear receptors. We conclude that the genetic programs specifying the identity and differentiation of arcuate POMC neurons are diverse and generate a heterogeneous repertoire of neuronal phenotypes early in development that continue to mature postnatally.
Collapse
Affiliation(s)
- Hui Yu
- Department of Molecular and Integrative Physiology, University of Michigan Medical SchoolAnn ArborUnited States
| | - Marcelo Rubinstein
- Department of Molecular and Integrative Physiology, University of Michigan Medical SchoolAnn ArborUnited States
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos AiresArgentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos AiresArgentina
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan Medical SchoolAnn ArborUnited States
| |
Collapse
|
19
|
Lu Y, Shi C, Jin X, He J, Yin Z. Domestication of farmed fish via the attenuation of stress responses mediated by the hypothalamus-pituitary-inter-renal endocrine axis. Front Endocrinol (Lausanne) 2022; 13:923475. [PMID: 35937837 PMCID: PMC9353172 DOI: 10.3389/fendo.2022.923475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022] Open
Abstract
Human-directed domestication of terrestrial animals traditionally requires thousands of years for breeding. The most prominent behavioral features of domesticated animals include reduced aggression and enhanced tameness relative to their wild forebears, and such behaviors improve the social tolerance of domestic animals toward both humans and crowds of their own species. These behavioral responses are primarily mediated by the hypothalamic-pituitary-adrenal (inter-renal in fish) (HPA/I) endocrine axis, which is involved in the rapid conversion of neuronal-derived perceptual information into hormonal signals. Over recent decades, growing evidence implicating the attenuation of the HPA/I axis during the domestication of animals have been identified through comprehensive genomic analyses of the paleogenomic datasets of wild progenitors and their domestic congeners. Compared with that of terrestrial animals, domestication of most farmed fish species remains at early stages. The present review focuses on the application of HPI signaling attenuation to accelerate the domestication and genetic breeding of farmed fish. We anticipate that deeper understanding of HPI signaling and its implementation in the domestication of farmed fish will benefit genetic breeding to meet the global demands of the aquaculture industry.
Collapse
Affiliation(s)
- Yao Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Chuang Shi
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Xia Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jiangyan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hainan Yazhou Bay Seed Laboratory, Sanya, China
- *Correspondence: Zhan Yin,
| |
Collapse
|
20
|
Singh PK, Lutfy K. The Role of Beta-Endorphin in Cocaine-Induced Conditioned Place Preference, Its Extinction, and Reinstatement in Male and Female Mice. Front Behav Neurosci 2021; 15:763336. [PMID: 34955777 PMCID: PMC8702804 DOI: 10.3389/fnbeh.2021.763336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/16/2021] [Indexed: 11/21/2022] Open
Abstract
Endogenous opioids have been implicated in cocaine reward. However, the role of each opioid peptide in this regard is unknown. Notably, the role of each peptide in extinction and reinstatement is not fully characterized. Thus, we assessed whether cocaine-induced conditioned place preference (CPP) and its extinction and reinstatement would be altered in the absence of beta-endorphin. We also examined if sex-related differences would exist in these processes. Male and female mice lacking beta-endorphin and their respective controls were tested for baseline place preference on day 1. On day 2, mice were treated with saline/cocaine (15 mg/kg) and confined to the vehicle- or drug-paired chamber for 30 min, respectively. In the afternoon, mice were treated with the alternate treatment and confined to the opposite chamber. Mice were then tested for CPP on day 3. Mice then received additional conditioning on this day as well as on day 4. Mice were then tested for CPP on day 5. Mice then received extinction training on day 9. On day 10, mice were tested for extinction and then reinstatement of CPP following a priming dose of cocaine (7.5 mg/kg). Male and female mice lacking beta-endorphin did not exhibit CPP following single conditioning with cocaine. On the other hand, only male mice lacking beta-endorphin failed to show CPP after repeated conditioning. Nonetheless, reinstatement of CPP was blunted in both male and female mice lacking beta-endorphin compared to controls. The present results suggest that beta-endorphin plays a functional role in cocaine-induced CPP and its reinstatement, and sex-related differences exist in the regulatory action of beta-endorphin on the acquisition but not reinstatement of cocaine CPP.
Collapse
Affiliation(s)
- Prableen K Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Kabirullah Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
21
|
Gupta A, Gullapalli S, Pan H, Ramos-Ortolaza DL, Hayward MD, Low MJ, Pintar JE, Devi LA, Gomes I. Regulation of Opioid Receptors by Their Endogenous Opioid Peptides. Cell Mol Neurobiol 2021; 41:1103-1118. [PMID: 33389463 PMCID: PMC8277103 DOI: 10.1007/s10571-020-01015-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022]
Abstract
Activation of μ, δ, and κ opioid receptors by endogenous opioid peptides leads to the regulation of many emotional and physiological responses. The three major endogenous opioid peptides, β-endorphin, enkephalins, and dynorphins result from the processing of three main precursors: proopiomelanocortin, proenkephalin, and prodynorphin. Using a knockout approach, we sought to determine whether the absence of endogenous opioid peptides would affect the expression or activity of opioid receptors in mice lacking either proenkephalin, β-endorphin, or both. Since gene knockout can lead to changes in the levels of peptides generated from related precursors by compensatory mechanisms, we directly measured the levels of Leu-enkephalin and dynorphin-derived peptides in the brain of animals lacking proenkephalin, β-endorphin, or both. We find that whereas the levels of dynorphin-derived peptides were relatively unaltered, the levels of Leu-enkephalin were substantially decreased compared to wild-type mice suggesting that preproenkephalin is the major source of Leu-enkephalin. This data also suggests that the lack of β-endorphin and/or proenkephalin does not lead to a compensatory change in prodynorphin processing. Next, we examined the effect of loss of the endogenous peptides on the regulation of opioid receptor levels and activity in specific regions of the brain. We also compared the receptor levels and activity in males and females and show that the lack of β-endorphin and/or proenkephalin leads to differential modulation of the three opioid receptors in a region- and gender-specific manner. These results suggest that endogenous opioid peptides are important modulators of the expression and activity of opioid receptors in the brain.
Collapse
Affiliation(s)
- Achla Gupta
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
| | - Srinivas Gullapalli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
- Emcure Pharmaceuticals, Mumbai, India
| | - Hui Pan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
- University of Southern California Medical Center, Los Angeles, CA, USA
| | - Dinah L Ramos-Ortolaza
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
- Pontifico Catholic Univ. Puerto Rico, Ponce, Puerto Rico
| | - Michael D Hayward
- Vollum Institute, Oregon Health Sciences University, Portland, OR, 97201, USA
- Invivotek, Trenton, NJ, USA
| | - Malcom J Low
- Vollum Institute, Oregon Health Sciences University, Portland, OR, 97201, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - John E Pintar
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA.
| | - Ivone Gomes
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA.
| |
Collapse
|
22
|
Kemény LV, Robinson KC, Hermann AL, Walker DM, Regan S, Yew YW, Lai YC, Theodosakis N, Rivera PD, Ding W, Yang L, Beyer T, Loh YHE, Lo JA, van der Sande AAJ, Sarnie W, Kotler D, Hsiao JJ, Su MY, Kato S, Kotler J, Bilbo SD, Chopra V, Salomon MP, Shen S, Hoon DSB, Asgari MM, Wakeman SE, Nestler EJ, Fisher DE. Vitamin D deficiency exacerbates UV/endorphin and opioid addiction. SCIENCE ADVANCES 2021; 7:7/24/eabe4577. [PMID: 34117054 PMCID: PMC8195487 DOI: 10.1126/sciadv.abe4577] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 04/26/2021] [Indexed: 06/12/2023]
Abstract
The current opioid epidemic warrants a better understanding of genetic and environmental factors that contribute to opioid addiction. Here we report an increased prevalence of vitamin D (VitD) deficiency in patients diagnosed with opioid use disorder and an inverse and dose-dependent association of VitD levels with self-reported opioid use. We used multiple pharmacologic approaches and genetic mouse models and found that deficiencies in VitD signaling amplify exogenous opioid responses that are normalized upon restoration of VitD signaling. Similarly, physiologic endogenous opioid analgesia and reward responses triggered by ultraviolet (UV) radiation are repressed by VitD signaling, suggesting that a feedback loop exists whereby VitD deficiency produces increased UV/endorphin-seeking behavior until VitD levels are restored by cutaneous VitD synthesis. This feedback may carry the evolutionary advantage of maximizing VitD synthesis. However, unlike UV exposure, exogenous opioid use is not followed by VitD synthesis (and its opioid suppressive effects), contributing to maladaptive addictive behavior.
Collapse
Affiliation(s)
- Lajos V Kemény
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathleen C Robinson
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea L Hermann
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deena M Walker
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Regan
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Yi Chun Lai
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Nicholas Theodosakis
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Phillip D Rivera
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA, USA
- Department of Biology, Hope College, Holland, MI, USA
| | - Weihua Ding
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liuyue Yang
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias Beyer
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yong-Hwee E Loh
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- USC Libraries Bioinformatics Services, University of Southern California, Los Angeles, CA, USA
| | - Jennifer A Lo
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anita A J van der Sande
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - William Sarnie
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Kotler
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer J Hsiao
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mack Y Su
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shinichiro Kato
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph Kotler
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Staci D Bilbo
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Vanita Chopra
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Matthew P Salomon
- Department of Translational Molecular Medicine, Division of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Shiqian Shen
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dave S B Hoon
- Department of Translational Molecular Medicine, Division of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Maryam M Asgari
- Department of Dermatology, Massachusetts General Hospital and Department of Population Medicine, Harvard Medical School, Boston, MA, USA
| | - Sarah E Wakeman
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Jaschke N, Pählig S, Pan YX, Hofbauer LC, Göbel A, Rachner TD. From Pharmacology to Physiology: Endocrine Functions of μ-Opioid Receptor Networks. Trends Endocrinol Metab 2021; 32:306-319. [PMID: 33676828 PMCID: PMC8035298 DOI: 10.1016/j.tem.2021.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 01/04/2023]
Abstract
The steady rise in opioid users and abusers has uncovered multiple detrimental health consequences of perturbed opioid receptor signaling, thereby creating the need to better understand the biology of these systems. Among endogenous opioid networks, μ-receptors have received special attention due to their unprecedented biological complexity and broad implications in homeostatic functions. Here, we review the origin, molecular biology, and physiology of endogenous opioids with a special focus on μ-opioid receptor networks within the endocrine system. Moreover, we summarize the current evidence supporting an involvement of the latter in regulating distinct endocrine functions. Finally, we combine these insights to present an integrated perspective on μ-opioid receptor biology and provide an outlook on future studies and unresolved questions in this field.
Collapse
Affiliation(s)
- Nikolai Jaschke
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| | - Sophie Pählig
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ying-Xian Pan
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Andy Göbel
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
24
|
Abrimian A, Kraft T, Pan YX. Endogenous Opioid Peptides and Alternatively Spliced Mu Opioid Receptor Seven Transmembrane Carboxyl-Terminal Variants. Int J Mol Sci 2021; 22:3779. [PMID: 33917474 PMCID: PMC8038826 DOI: 10.3390/ijms22073779] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 12/20/2022] Open
Abstract
There exist three main types of endogenous opioid peptides, enkephalins, dynorphins and β-endorphin, all of which are derived from their precursors. These endogenous opioid peptides act through opioid receptors, including mu opioid receptor (MOR), delta opioid receptor (DOR) and kappa opioid receptor (KOR), and play important roles not only in analgesia, but also many other biological processes such as reward, stress response, feeding and emotion. The MOR gene, OPRM1, undergoes extensive alternative pre-mRNA splicing, generating multiple splice variants or isoforms. One type of these splice variants, the full-length 7 transmembrane (TM) Carboxyl (C)-terminal variants, has the same receptor structures but contains different intracellular C-terminal tails. The pharmacological functions of several endogenous opioid peptides through the mouse, rat and human OPRM1 7TM C-terminal variants have been considerably investigated together with various mu opioid ligands. The current review focuses on the studies of these endogenous opioid peptides and summarizes the results from early pharmacological studies, including receptor binding affinity and G protein activation, and recent studies of β-arrestin2 recruitment and biased signaling, aiming to provide new insights into the mechanisms and functions of endogenous opioid peptides, which are mediated through the OPRM1 7TM C-terminal splice variants.
Collapse
Affiliation(s)
| | | | - Ying-Xian Pan
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (A.A.); (T.K.)
| |
Collapse
|
25
|
Robinson KC, Kemény LV, Fell GL, Hermann AL, Allouche J, Ding W, Yekkirala A, Hsiao JJ, Su MY, Theodosakis N, Kozak G, Takeuchi Y, Shen S, Berenyi A, Mao J, Woolf CJ, Fisher DE. Reduced MC4R signaling alters nociceptive thresholds associated with red hair. SCIENCE ADVANCES 2021; 7:eabd1310. [PMID: 33811065 PMCID: PMC11057701 DOI: 10.1126/sciadv.abd1310] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/16/2021] [Indexed: 06/12/2023]
Abstract
Humans and mice with natural red hair have elevated basal pain thresholds and an increased sensitivity to opioid analgesics. We investigated the mechanisms responsible for higher nociceptive thresholds in red-haired mice resulting from a loss of melanocortin 1 receptor (MC1R) function and found that the increased thresholds are melanocyte dependent but melanin independent. MC1R loss of function decreases melanocytic proopiomelanocortin transcription and systemic melanocyte-stimulating hormone (MSH) levels in the plasma of red-haired (Mc1re/e ) mice. Decreased peripheral α-MSH derepresses the central opioid tone mediated by the opioid receptor OPRM1, resulting in increased nociceptive thresholds. We identified MC4R as the MSH-responsive receptor that opposes OPRM1 signaling and the periaqueductal gray area in the brainstem as a central area of opioid/melanocortin antagonism. This work highlights the physiologic role of melanocytic MC1R and circulating melanocortins in the regulation of nociception and provides a mechanistic framework for altered opioid signaling and pain sensitivity in red-haired individuals.
Collapse
Affiliation(s)
- Kathleen C Robinson
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Lajos V Kemény
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Gillian L Fell
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Andrea L Hermann
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Doctoral School of Clinical Medicine, University of Szeged, Szeged 6720, Hungary
| | - Jennifer Allouche
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Weihua Ding
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Ajay Yekkirala
- FM Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer J Hsiao
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Mack Y Su
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Nicholas Theodosakis
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Gabor Kozak
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
- University Neurology Hospital and Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Yuichi Takeuchi
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
- Neurocybernetics Excellence Center, University of Szeged, 10 Dom sqr, Szeged 6720, Hungary
- Department of Physiology, Osaka City University Graduate School of Medicine, 1-4-3, Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Shiqian Shen
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Antal Berenyi
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
- Neurocybernetics Excellence Center, University of Szeged, 10 Dom sqr, Szeged 6720, Hungary
- Neuroscience Institute, New York University, New York City, NY 10016, USA
- HCEMM-USZ Magnetotherapeutics Research Group, University of Szeged, 10 Dom sqr, Szeged 6720, Hungary
| | - Jianren Mao
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Clifford J Woolf
- FM Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
26
|
Quarta C, Claret M, Zeltser LM, Williams KW, Yeo GSH, Tschöp MH, Diano S, Brüning JC, Cota D. POMC neuronal heterogeneity in energy balance and beyond: an integrated view. Nat Metab 2021; 3:299-308. [PMID: 33633406 PMCID: PMC8085907 DOI: 10.1038/s42255-021-00345-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023]
Abstract
Hypothalamic AgRP and POMC neurons are conventionally viewed as the yin and yang of the body's energy status, since they act in an opposite manner to modulate appetite and systemic energy metabolism. However, although AgRP neurons' functions are comparatively well understood, a unifying theory of how POMC neuronal cells operate has remained elusive, probably due to their high level of heterogeneity, which suggests that their physiological roles might be more complex than initially thought. In this Perspective, we propose a conceptual framework that integrates POMC neuronal heterogeneity with appetite regulation, whole-body metabolic physiology and the development of obesity. We highlight emerging evidence indicating that POMC neurons respond to distinct combinations of interoceptive signals and food-related cues to fine-tune divergent metabolic pathways and behaviours necessary for survival. The new framework we propose reflects the high degree of developmental plasticity of this neuronal population and may enable progress towards understanding of both the aetiology and treatment of metabolic disorders.
Collapse
Affiliation(s)
- Carmelo Quarta
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, INSERM U1215, Bordeaux, France
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER), Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Lori M Zeltser
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| | - Sabrina Diano
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY, USA
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- National Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Daniela Cota
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, INSERM U1215, Bordeaux, France.
| |
Collapse
|
27
|
Cioato SG, Medeiros LF, Lopes BC, de Souza A, Medeiros HR, Assumpção JAF, Caumo W, Roesler R, Torres ILS. Antinociceptive and neurochemical effects of a single dose of IB-MECA in chronic pain rat models. Purinergic Signal 2020; 16:573-584. [PMID: 33161497 PMCID: PMC7855191 DOI: 10.1007/s11302-020-09751-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 10/18/2020] [Indexed: 12/31/2022] Open
Abstract
This study aimed to evaluate the effect of a single administration of IB-MECA, an A3 adenosine receptor agonist, upon the nociceptive response and central biomarkers of rats submitted to chronic pain models. A total of 136 adult male Wistar rats were divided into two protocols: (1) chronic inflammatory pain (CIP) using complete Freund's adjuvant and (2) neuropathic pain (NP) by chronic constriction injury of the sciatic nerve. Thermal and mechanical hyperalgesia was measured using von Frey (VF), Randal-Selitto (RS), and hot plate (HP) tests. Rats were treated with a single dose of IB-MECA (0.5 μmol/kg i.p.), a vehicle (dimethyl sulfoxide-DMSO), or positive control (morphine, 5 mg/kg i.p.). Interleukin 1β (IL-1β), brain-derived neurotrophic factor (BDNF), and nerve growth factor (NGF) levels were measured in the brainstem and spinal cord using enzyme-linked immunosorbent assay (ELISA). The establishment of the chronic pain (CIP or NP) model was observed 14 days after induction by a decreased nociceptive threshold in all three tests (GEE, P < 0.05). The antinociceptive effect of a single dose of IB-MECA was observed in both chronic pain models, but this was more effective in NP model. There was an increase in IL-1β levels promoted by CIP. NP model promoted increase in the brainstem BDNF levels, which was reversed by IB-MECA.
Collapse
Affiliation(s)
- Stefania Giotti Cioato
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Unidade de Experimentação Animal, Grupo de Pesquisa e Pós-Graduação, HCPA, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica, Instituto de Ciências Básicas da Saúde (ICBS), UFRGS, Porto Alegre, RS, Brazil
| | - Liciane Fernandes Medeiros
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Unidade de Experimentação Animal, Grupo de Pesquisa e Pós-Graduação, HCPA, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica, Instituto de Ciências Básicas da Saúde (ICBS), UFRGS, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Saúde e Desenvolvimento Humano, Universidade La Salle, Canoas, RS, Brazil
| | - Bettega Costa Lopes
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Unidade de Experimentação Animal, Grupo de Pesquisa e Pós-Graduação, HCPA, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, ICBS, UFRGS, Porto Alegre, RS, Brazil
| | - Andressa de Souza
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Programa de Pós-Graduação em Saúde e Desenvolvimento Humano, Universidade La Salle, Canoas, RS, Brazil
| | - Helouise Richardt Medeiros
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Unidade de Experimentação Animal, Grupo de Pesquisa e Pós-Graduação, HCPA, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Medicina, Ciências Médicas, UFRGS, Porto Alegre, RS, Brazil
| | - José Antônio Fagundes Assumpção
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil
- Unidade de Experimentação Animal, Grupo de Pesquisa e Pós-Graduação, HCPA, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica, Instituto de Ciências Básicas da Saúde (ICBS), UFRGS, Porto Alegre, RS, Brazil
| | - Wolnei Caumo
- Programa de Pós-Graduação em Medicina, Ciências Médicas, UFRGS, Porto Alegre, RS, Brazil
| | - Rafael Roesler
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica, Instituto de Ciências Básicas da Saúde (ICBS), UFRGS, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Medicina, Ciências Médicas, UFRGS, Porto Alegre, RS, Brazil
- Cancer and Neurobiology Laboratory, Experimental Research Center, HCPA, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-007, Brazil.
- Unidade de Experimentação Animal, Grupo de Pesquisa e Pós-Graduação, HCPA, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica, Instituto de Ciências Básicas da Saúde (ICBS), UFRGS, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação em Medicina, Ciências Médicas, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
28
|
Transcranial Direct Current Stimulation (tDCS) Induces Analgesia in Rats with Neuropathic Pain and Alcohol Abstinence. Neurochem Res 2020; 45:2653-2663. [DOI: 10.1007/s11064-020-03116-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/31/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023]
|
29
|
Machelska H, Celik MÖ. Immune cell-mediated opioid analgesia. Immunol Lett 2020; 227:48-59. [PMID: 32814155 DOI: 10.1016/j.imlet.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022]
Abstract
Pathological pain is regulated by a balance between pro-algesic and analgesic mechanisms. Interactions between opioid peptide-producing immune cells and peripheral sensory neurons expressing opioid receptors represent a powerful intrinsic pain control in animal models and in humans. Therefore, treatments based on general suppression of immune responses have been mostly unsuccessful. It is highly desirable to develop strategies that specifically promote neuro-immune communication mediated by opioids. Promising examples include vaccination-based recruitment of opioid-containing leukocytes to painful tissue and the local reprogramming of pro-algesic immune cells into analgesic cells producing and secreting high amounts of opioid peptides. Such approaches have the potential to inhibit pain at its origin and be devoid of central and systemic side effects of classical analgesics. In support of these concepts, in this article, we describe the functioning of peripheral opioid receptors, migration of opioid-producing immune cells to inflamed tissue, opioid peptide release, and the consequent pain relief. Conclusively, we provide clinical evidence and discuss therapeutic opportunities and challenges associated with immune cell-mediated peripheral opioid analgesia.
Collapse
Affiliation(s)
- Halina Machelska
- Department of Experimental Anesthesiology, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany.
| | - Melih Ö Celik
- Department of Experimental Anesthesiology, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
30
|
Fricker LD, Margolis EB, Gomes I, Devi LA. Five Decades of Research on Opioid Peptides: Current Knowledge and Unanswered Questions. Mol Pharmacol 2020; 98:96-108. [PMID: 32487735 PMCID: PMC7330675 DOI: 10.1124/mol.120.119388] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
In the mid-1970s, an intense race to identify endogenous substances that activated the same receptors as opiates resulted in the identification of the first endogenous opioid peptides. Since then, >20 peptides with opioid receptor activity have been discovered, all of which are generated from three precursors, proenkephalin, prodynorphin, and proopiomelanocortin, by sequential proteolytic processing by prohormone convertases and carboxypeptidase E. Each of these peptides binds to all three of the opioid receptor types (μ, δ, or κ), albeit with differing affinities. Peptides derived from proenkephalin and prodynorphin are broadly distributed in the brain, and mRNA encoding all three precursors are highly expressed in some peripheral tissues. Various approaches have been used to explore the functions of the opioid peptides in specific behaviors and brain circuits. These methods include directly administering the peptides ex vivo (i.e., to excised tissue) or in vivo (in animals), using antagonists of opioid receptors to infer endogenous peptide activity, and genetic knockout of opioid peptide precursors. Collectively, these studies add to our current understanding of the function of endogenous opioids, especially when similar results are found using different approaches. We briefly review the history of identification of opioid peptides, highlight the major findings, address several myths that are widely accepted but not supported by recent data, and discuss unanswered questions and future directions for research. SIGNIFICANCE STATEMENT: Activation of the opioid receptors by opiates and synthetic drugs leads to central and peripheral biological effects, including analgesia and respiratory depression, but these may not be the primary functions of the endogenous opioid peptides. Instead, the opioid peptides play complex and overlapping roles in a variety of systems, including reward pathways, and an important direction for research is the delineation of the role of individual peptides.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Elyssa B Margolis
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Ivone Gomes
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| | - Lakshmi A Devi
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.); Department of Neurology, UCSF Weill Institute for Neurosciences, San Francisco, California (E.B.M.); and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (I.G., L.A.D.)
| |
Collapse
|
31
|
Nakamoto K, Taniguchi A, Tokuyama S. Changes in opioid receptors, opioid peptides and morphine antinociception in mice subjected to early life stress. Eur J Pharmacol 2020; 881:173173. [PMID: 32511976 DOI: 10.1016/j.ejphar.2020.173173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 02/09/2023]
Abstract
Recent studies have shown that the endogenous opioid system is considerably affected by early life stress such as child abuse. Here, we investigated whether early life stress changes the endogenous opioid receptors and their peptides, and if such stress impacts morphine antinociception. We used mice affected by maternal separation and social isolation (MSSI) as an early life stress model. In the tail-flick test, 10-week-old MSSI mice showed a significant decrease in morphine antinociception compared to age-matched control mice. The number of c-Fos-positive cells increased in the periaqueductal gray (PAG), nucleus accumbens, and thalamus of control mice after the morphine injections, whereas hardly any positive cells were detected in the same areas of MSSI mice. The expression of μ- and κ-opioid receptor (MOR and KOR, respectively) messenger RNA (mRNA) was significantly decreased in the PAG of MSSI mice, whereas KOR expression was significantly increased in the amygdala of MSSI mice. The expression of δ-opioid receptor (DOR) mRNA was significantly reduced in the PAG and rostral ventromedial medulla of MSSI mice compared to control mice. Moreover, the lack of morphine antinociception was observed in 18-week-old MSSI mice. Our findings suggest that the supraspinal opioid system may be affected by early life stress exposure, and that this exposure may impact morphine antinociception.
Collapse
Affiliation(s)
- Kazuo Nakamoto
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe, 650-8586, Japan
| | - Ayaka Taniguchi
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe, 650-8586, Japan
| | - Shogo Tokuyama
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe, 650-8586, Japan.
| |
Collapse
|
32
|
Shi C, Lu Y, Zhai G, Huang J, Shang G, Lou Q, Li D, Jin X, He J, Du Z, Gui J, Yin Z. Hyperandrogenism in POMCa-deficient zebrafish enhances somatic growth without increasing adiposity. J Mol Cell Biol 2020; 12:291-304. [PMID: 31237951 PMCID: PMC7232124 DOI: 10.1093/jmcb/mjz053] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/17/2019] [Accepted: 05/17/2019] [Indexed: 01/29/2023] Open
Abstract
The endocrine regulatory roles of the hypothalamic-pituitary-adrenocortical axis on anxiety-like behavior and metabolic status have been found throughout animal taxa. However, the precise effects of the balancing adrenal corticosteroid biosynthesis under the influence of adrenocorticotrophic hormone (ACTH), a pro-opiomelanocortin (POMC)-derived peptide, on animal energy expenditure and somatic growth remain unknown. POMC has also been identified as one of the candidate loci for polycystic ovary syndrome, which features hyperandrogenism and some prevalence of obesity in patients. Here we show that zebrafish lacking functional POMCa exhibit similar phenotypes of stress response and body weight gain but not obesity as observed in mammalian models. In contrast with the impaired anorexigenic signaling cascade of melanocyte-stimulating hormones and leptin, which are responsible for their obesity-prone weight gain observed in various pomc mutant mammals, analyses with our pomca mutant series indicate that ACTH is the key regulator for the phenotype with enhanced somatic growth without obesity in pomca-deficient zebrafish. Hypocortisolism associated with hyperandrogenism has been observed in the pomca-deficient zebrafish, with enhanced activation of mammalian target of rapamycin complex 1; reutilization of amino acids and fatty acid β-oxidation are observed in the muscle tissue of the pomca-deficient fish. After reducing hyperandrogenism by crossing our pomca mutant fish with a cyp17a1-deficient background, the phenotype of enhanced somatic growth in pomca-deficient fish was no longer observed. Thus, our work also demonstrated that the role of POMCa in stress response seems to be conserved in vertebrates, whereas its effect on adipostasis is unique to teleosts.
Collapse
Affiliation(s)
- Chuang Shi
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Zhai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jianfei Huang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guohui Shang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiyong Lou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Dongliang Li
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xia Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jiangyan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zhenyu Du
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jianfang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing 100864, China
| |
Collapse
|
33
|
Bravo L, Llorca-Torralba M, Suárez-Pereira I, Berrocoso E. Pain in neuropsychiatry: Insights from animal models. Neurosci Biobehav Rev 2020; 115:96-115. [PMID: 32437745 DOI: 10.1016/j.neubiorev.2020.04.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 04/11/2020] [Accepted: 04/23/2020] [Indexed: 02/08/2023]
Abstract
Pain is the most common symptom reported in clinical practice, meaning that it is associated with many pathologies as either the origin or a consequence of other illnesses. Furthermore, pain is a complex emotional and sensorial experience, as the correspondence between pain and body damage varies considerably. While these issues are widely acknowledged in clinical pain research, until recently they have not been extensively considered when exploring animal models, important tools for understanding pain pathophysiology. Interestingly, chronic pain is currently considered a risk factor to suffer psychiatric disorders, mainly stress-related disorders like anxiety and depression. Conversely, pain appears to be altered in many psychiatric disorders, such as depression, anxiety and schizophrenia. Thus, pain and psychiatric disorders have been linked in epidemiological and clinical terms, although the neurobiological mechanisms involved in this pathological bidirectional relationship remain unclear. Here we review the evidence obtained from animal models about the co-morbidity of pain and psychiatric disorders, placing special emphasis on the different dimensions of pain.
Collapse
Affiliation(s)
- Lidia Bravo
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Suárez-Pereira
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Neuropsychopharmacology and Psychobiology Research Group, Department of Psychology, University of Cádiz, 11510 Puerto Real, Cádiz, Spain.
| |
Collapse
|
34
|
Ströher R, de Oliveira C, Costa Lopes B, da Silva LS, Regner GG, Richardt Medeiros H, de Macedo IC, Caumo W, Torres ILS. Maternal deprivation alters nociceptive response in a gender-dependent manner in rats. Int J Dev Neurosci 2019; 76:25-33. [PMID: 31071409 DOI: 10.1016/j.ijdevneu.2019.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 04/07/2019] [Accepted: 05/03/2019] [Indexed: 12/27/2022] Open
Abstract
The present study aimed at investigating both the early and long-term effects of maternal deprivation as well as gender on neuromotor reflexes, anxiety behavior and thermal nociceptive responses. A total of 64 Wistar rats pups (32 males, 32 females) were utilized and were deprived of their mother for 3 h/daily, from postnatal day 1 (P1) until P10. Successively, animals were divided into 2 groups: control group (C) - pups no subjected to intervention; and the maternal-deprived group (MD): pups subjected to maternal deprivation. The neuromotor reflexes were evaluated through the righting reflex and negative geotaxis tests; the exploratory behavior by open field test (OFT); the anxiety-like behavior by elevated plus-maze test (EPM); the thermal nociceptive responses byhot plate (HP) and tail-flick (TFL) tests. All the animals subjected to maternal deprivation showed a delayed reflex response at P8 in the negative geotaxis test. In contrast, the OFT at P20 identified an effect of gender on the outer crossings and grooming as well as an interaction between gender and maternal deprivation on latency. Additionally, effect of maternal deprivation in the open and closed arms as well as gender effect in the protected head-dipping (PHD) and non-protected head-dipping (NPHD) were observed at P20 (EPM). In contrast, there were a gender effect on latency and an interaction between gender and maternal deprivation on rearing at P42. Moreover, in nociceptive tests was observed an analgesic effect induced by maternal deprivation; however, in the TFL test, only deprived females showed this effect. Surprisingly, only control animals presented an ontogeny nociceptive effect in the HP testat P21 and P43, which may be related to an increase in the inhibitory nociceptive pathways throughout life. In this way, we suggest maternal deprivation to be able to anticipate the maturation of the inhibitory nociceptive pathway. In conclusion, maternal deprivation induced a delayed reflex response at P8 and altered the anxiety and nociceptive behaviors according to the time after exposure to this stressor, in a gender-specific manner.
Collapse
Affiliation(s)
- Roberta Ströher
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica-Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil
| | - Carla de Oliveira
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bettega Costa Lopes
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas, Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lisiane Santos da Silva
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gabriela Gregory Regner
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica-Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil
| | - Helouise Richardt Medeiros
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Isabel Cristina de Macedo
- Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil.,Universidade Federal do Pampa, São Gabriel, RS, Brazil
| | - Wolnei Caumo
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Programa de Pós-Graduação em Ciências Biológicas, Farmacologia e Terapêutica-Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Departamento de Farmacologia, Instituto de CiênciasBásicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Laboratório de Farmacologia da Dor e Neuromodulação, Investigações Pré-clínicas, Departamento de Farmacologia, ICBS, UFRGS, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas, Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
35
|
Nentwig TB, Wilson DE, Rhinehart EM, Grisel JE. Sex differences in binge-like EtOH drinking, corticotropin-releasing hormone and corticosterone: effects of β-endorphin. Addict Biol 2019; 24:447-457. [PMID: 29424043 PMCID: PMC6082742 DOI: 10.1111/adb.12610] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/13/2018] [Accepted: 01/16/2018] [Indexed: 01/08/2023]
Abstract
Binge drinking is an increasingly common pattern of risky use associated with numerous health problems, including alcohol use disorders. Because low basal plasma levels of β-endorphin (β-E) and an increased β-E response to alcohol are evident in genetically at-risk human populations, this peptide is thought to contribute to the susceptibility for disordered drinking. Animal models suggest that the effect of β-E on consumption may be sex-dependent. Here, we studied binge-like EtOH consumption in transgenic mice possessing varying levels of β-E: wild-type controls with 100% of the peptide (β-E +/+), heterozygous mice constitutively modified to possess 50% of wild-type levels (β-E +/-) and mice entirely lacking the capacity to synthesize β-E (-/-). These three genotypes and both sexes were evaluated in a 4-day, two-bottle choice, drinking in the dark paradigm with limited access to 20% EtOH. β-E deficiency determined sexually divergent patterns of drinking in that β-E -/- female mice drank more than their wild-type counterparts, an effect not observed in male mice. β-E -/- female mice also displayed elevated basal anxiety, plasma corticosterone and corticotropin-releasing hormone mRNA in the extended amygdala, and all of these were normalized by EtOH self-administration. These data suggest that a heightened risk for excessive EtOH consumption in female mice is related to the drug's ability to ameliorate an overactive anxiety/stress-like state. Taken together, our study highlights a critical impact of sex on neuropeptide regulation of EtOH consumption.
Collapse
Affiliation(s)
- Todd B. Nentwig
- Department of Psychology, Neuroscience ProgramBucknell UniversityLewisburgPAUSA
| | | | | | - Judith E. Grisel
- Department of Psychology, Neuroscience ProgramBucknell UniversityLewisburgPAUSA
| |
Collapse
|
36
|
Harno E, Gali Ramamoorthy T, Coll AP, White A. POMC: The Physiological Power of Hormone Processing. Physiol Rev 2019; 98:2381-2430. [PMID: 30156493 DOI: 10.1152/physrev.00024.2017] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pro-opiomelanocortin (POMC) is the archetypal polypeptide precursor of hormones and neuropeptides. In this review, we examine the variability in the individual peptides produced in different tissues and the impact of the simultaneous presence of their precursors or fragments. We also discuss the problems inherent in accurately measuring which of the precursors and their derived peptides are present in biological samples. We address how not being able to measure all the combinations of precursors and fragments quantitatively has affected our understanding of the pathophysiology associated with POMC processing. To understand how different ratios of peptides arise, we describe the role of the pro-hormone convertases (PCs) and their tissue specificities and consider the cellular processing pathways which enable regulated secretion of different peptides that play crucial roles in integrating a range of vital physiological functions. In the pituitary, correct processing of POMC peptides is essential to maintain the hypothalamic-pituitary-adrenal axis, and this processing can be disrupted in POMC-expressing tumors. In hypothalamic neurons expressing POMC, abnormalities in processing critically impact on the regulation of appetite, energy homeostasis, and body composition. More work is needed to understand whether expression of the POMC gene in a tissue equates to release of bioactive peptides. We suggest that this comprehensive view of POMC processing, with a focus on gaining a better understanding of the combination of peptides produced and their relative bioactivity, is a necessity for all involved in studying this fascinating physiological regulatory phenomenon.
Collapse
Affiliation(s)
- Erika Harno
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| | - Thanuja Gali Ramamoorthy
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| | - Anthony P Coll
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| | - Anne White
- Division of Diabetes, Endocrinology and Gastrointestinal Sciences, Faculty of Biology, Medicine and Health, University of Manchester , Manchester , United Kingdom ; and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science , Cambridge , United Kingdom
| |
Collapse
|
37
|
Gong LL, Yang S, Liu H, Zhang W, Ren LL, Han FF, Lv YL, Wan ZR, Liu LH. Anti-nociceptive and anti-inflammatory potentials of Akebia saponin D. Eur J Pharmacol 2018; 845:85-90. [PMID: 30508505 DOI: 10.1016/j.ejphar.2018.11.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/28/2018] [Accepted: 11/28/2018] [Indexed: 11/24/2022]
Abstract
Akebia saponin D, which is originates from Dipsacus asper Wall, has been used as a tonic, an analgesic and anti-inflammatory agent for the therapy of low back pain, rheumatic arthritis, traumatic hematoma, habitual abortion and bone fractures in traditional Chinese medicine. However, the anti-nociceptive and anti-inflammatory activity and mechanism of Akebia saponin D has been rarely reported. The aim of this study was to investigate the anti-nociceptive and anti-inflammatory activity of Akebia saponin D and to assess its possible mechanism. The anti-nociceptive effect was measured by formalin test, hot plate, and acetic acid-induced writhing in mice while the anti-inflammatory effect was measured by carrageenan induced paw edema test, xylene-induced ear swelling and acetic acid-induced vascular permeability in mice and rats. Furthermore, anti-inflammatory effect was also measured in vitro using LPS-induced RAW 264.7 cells. Our results demonstrated that Akebia saponin D dose-dependently decreased the licking time in the formalin test, delayed the reaction time of mice to the hot plate, and inhibited acetic acid-induced writhing. Treatment of Akebia saponin D attenuated the carrageenan induced paw edema in rats, inhibited the mouse ear swelling, and decreased Evans blue concentration in acetic acid induced vascular permeability test, revealing its strong anti-inflammatory effect. Akebia saponin D significantly decreased NO production and iNOS expression. Our results indicate that Akebia saponin D has anti-nociceptive and anti-inflammatory effects. It will provide experimental evidences for the use of Akebia saponin D and can be used to develop a therapeutic drug against pain and inflammation related diseases.
Collapse
Affiliation(s)
- Li-Li Gong
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - Song Yang
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - He Liu
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - Wen Zhang
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - Lu-Lu Ren
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - Fei-Fei Han
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - Ya-Li Lv
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - Zi-Rui Wan
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China
| | - Li-Hong Liu
- Beijing Chao-Yang Hospital Affiliated with Beijing Capital Medical University, Beijing, China.
| |
Collapse
|
38
|
Rhinehart EM, Nentwig TB, Wilson DE, Leonard KT, Chaney BN, Grisel JE. Sex and β-Endorphin Influence the Effects of Ethanol on Limbic Gabra2 Expression in a Mouse Binge Drinking Model. Front Genet 2018; 9:567. [PMID: 30555510 PMCID: PMC6281685 DOI: 10.3389/fgene.2018.00567] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/06/2018] [Indexed: 11/13/2022] Open
Abstract
Binge drinking is a widespread problem linked to increased risk for alcohol-related complications, including development of alcohol use disorders. In the last decade, binge drinking has increased significantly, specifically in women. Clinically, sexually dimorphic effects of alcohol are well-characterized, however, the underlying mechanisms for these dimorphisms in the physiological and behavioral effects of alcohol are poorly understood. Among its many effects, alcohol consumption reduces anxiety via the inhibitory neurotransmitter GABA, most likely acting upon receptors containing the α-2 subunit (Gabra2). Previous research from our laboratory indicates that female mice lacking the endogenous opioid peptide β-endorphin (βE) have an overactive stress axis and enhanced anxiety-like phenotype, coupled with increased binge-like alcohol consumption. Because βE works via GABA signaling to reduce anxiety, we sought to determine whether sexually dimorphic binge drinking behavior in βE deficient mice is coupled with differences in CNS Gabra2 expression. To test this hypothesis, we used βE knock-out mice in a "drinking in the dark" model where adult male and female C57BL/6J controls (βE +/+) and βE deficient (βE -/-; B6.129S2-Pomctm1Low/J) mice were provided with one bottle of 20% ethanol (EtOH) and one of water (EtOH drinkers) or two bottles of water (water drinkers) 3 h into the dark cycle for four consecutive days. Following a binge test on day 4, limbic tissue was collected and frozen for subsequent qRT-PCR analysis of Gabra2 mRNA expression. Water-drinking βE +/+ females expressed more Gabra2 in central nucleus of the amygdala and the bed nucleus of the stria terminalis than males, but this sex difference was absent in the βE -/- mice. Genotype alone had no effect on alcohol consumption or drug-induced increase in Gabra2 expression. In contrast, βE expression had bi-directional effects in females: in wildtypes, Gabra2 mRNA was reduced by binge EtOH consumption, while EtOH increased expression in βE -/- females to levels commensurate with drug-naïve βE +/+ females. These results support the contention that βE plays a role in sexually dimorphic binge-like EtOH consumption, perhaps through differential expression of GABAA α2 subunits in limbic structures known to play key roles in the regulation of stress and anxiety.
Collapse
Affiliation(s)
- Erin M Rhinehart
- Department of Biology, Susquehanna University, Selinsgrove, PA, United States
| | - Todd B Nentwig
- Department of Psychology, Neuroscience Program, Bucknell University, Lewisburg, PA, United States
| | - Diane E Wilson
- Department of Biology, Susquehanna University, Selinsgrove, PA, United States
| | - Kiarah T Leonard
- Department of Psychology, Neuroscience Program, Bucknell University, Lewisburg, PA, United States
| | - Bernie N Chaney
- Department of Psychology, Neuroscience Program, Bucknell University, Lewisburg, PA, United States
| | - Judith E Grisel
- Department of Psychology, Neuroscience Program, Bucknell University, Lewisburg, PA, United States
| |
Collapse
|
39
|
Scarabelot VL, de Oliveira C, Medeiros LF, de Macedo IC, Cioato SG, Adachi LNS, Paz AH, de Souza A, Caumo W, Torres ILS. Transcranial direct-current stimulation reduces nociceptive behaviour in an orofacial pain model. J Oral Rehabil 2018; 46:40-50. [PMID: 30281821 DOI: 10.1111/joor.12726] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 09/17/2018] [Accepted: 09/26/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Transcranial direct-current stimulation (tDCS) is a noninvasive method of brain stimulation suggested as a therapeutic tool for pain and is related to the reversal of maladaptive plasticity associated with chronic pain. OBJECTIVES This study investigated the effect of tDCS, a non-pharmacological therapy, on local mechanical hyperalgesia, and remote thermal hyperalgesia in rats submitted to orofacial inflammatory pain model, by facial von Frey and hot plate tests, respectively. In addition, we evaluated levels of BDNF, NGF, IL-10 and IL-6 in the brainstem and blood serum of these animals at 24 hours and 7 days after the end of tDCS treatment. METHODS Rats were subjected to temporomandibular joint pain and treated with tDCS. The animals were divided into control, pain and pain + treatment groups. Mechanical and thermal hyperalgesia were evaluated at baseline, 7 days after administration of complete Freund's adjuvant, and immediately, 24 hours, and 7 days after the tDCS treatment. Neuroimmunomodulators levels were determined by ELISA. Statistical analyses were performed by (GEE)/Bonferroni (behavioural tests), three-way ANOVA/SNK (neurochemical tests) and Kruskal-Wallis (histological analysis). RESULTS Transcranial direct-current stimulation reduced mechanical and thermal hyperalgesia (P < 0.01). We observed interaction between factors (pain and treatment) increasing brainstem BDNF (P < 0.01) and NGF (P < 0.05) levels. Furthermore, we found an increase in IL-6 and IL-10 levels in the brainstem at 24 hours and 7 days after tDCS, respectively. CONCLUSION We showed that tDCS reduces thermal and mechanical hyperalgesia induced by orofacial pain until 7 days after treatment. These findings demonstrate that tDCS was effective in the control of orofacial inflammatory pain.
Collapse
Affiliation(s)
- Vanessa L Scarabelot
- Post-Graduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carla de Oliveira
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Liciane F Medeiros
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Isabel C de Macedo
- Post-Graduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Stefania G Cioato
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lauren Naomi S Adachi
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Helena Paz
- Morphological Sciences Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andressa de Souza
- Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Wolnei Caumo
- Department of Surgery in Medical School, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Iraci L S Torres
- Post-Graduate Program in Biological Sciences: Physiology, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Pain Pharmacology and Neuromodulation: Pre-clinical Research, Pharmacology Department, Health Basic Sciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
40
|
Poznański P, Lesniak A, Bujalska-Zadrozny M, Strzemecka J, Sacharczuk M. Bidirectional selection for high and low stress-induced analgesia affects G-protein activity. Neuropharmacology 2018; 144:37-42. [PMID: 30326238 DOI: 10.1016/j.neuropharm.2018.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/03/2018] [Accepted: 10/12/2018] [Indexed: 11/19/2022]
Abstract
Mice selected for high (HA) and low (LA) swim stress-induced analgesia (SSIA) are a unique model for studying the genetic background of this phenomenon. HA and LA miceshow substantial differences in the magnitude of the antinociceptive response to stress and when treated with exogenous opioids. However, the direct cause underplaying this distinctive feature has not yet been identified. The current study was designed to investigate the possibility that disturbances in G-protein signaling could explain the divergent response to opioid agonists. Supraspinal and spinal opioid sensitivity was assessed in vivo with intraperitoneal morphine and subsequent thermal stimulus exposure. The level of opioid receptor-mediated G-protein activation was investigated by means of DAMGO and morphine-stimulated [35S]GTPγS assay in the brain and spinal cord homogenates from HA and LA mice. Morphine (3-249 μmol/kg, i.p) was over 6 - and 3 - times more potent in HA than LA mice in the hot plate and tail-flick assays, respectively. Additionally, HA mice showed elevated β - endorphin levels in the brain. Enhanced efficacy of agonist-stimulated [35S]GTPγS binding was detected in opioid receptor-rich limbic regions of HA mice like the hypothalamus and hippocampus. Increased G-protein activity also emerged in the thalamus, periaqueductal gray matter and prefrontal cortex. In conclusion, the magnitude of the antinociceptive response to opioids in HA and LA mice is correlated with alterations in G-protein activation in brain regions responsible for integration and descending modulation of nociceptive information as well as at sites governing the emotional response to stressful stimuli.
Collapse
Affiliation(s)
- Piotr Poznański
- Department of Genomics, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Anna Lesniak
- Faculty of Pharmacy with the Laboratory Medicine Division, Department of Pharmacodynamics, Medical University of Warsaw, Centre for Preclinical Research and Technology, Warsaw, Poland
| | - Magdalena Bujalska-Zadrozny
- Faculty of Pharmacy with the Laboratory Medicine Division, Department of Pharmacodynamics, Medical University of Warsaw, Centre for Preclinical Research and Technology, Warsaw, Poland
| | - Joanna Strzemecka
- Institute of Health Sciences, Pope John Paul II State School of Higher Education, Biala Podlaska, Poland
| | - Mariusz Sacharczuk
- Faculty of Pharmacy with the Laboratory Medicine Division, Department of Pharmacodynamics, Medical University of Warsaw, Centre for Preclinical Research and Technology, Warsaw, Poland; Department of Genomics, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, Poland; Department of Internal Medicine, Hypertension and Vascular Diseases, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
41
|
Wu HY, Mao XF, Tang XQ, Ali U, Apryani E, Liu H, Li XY, Wang YX. Spinal interleukin-10 produces antinociception in neuropathy through microglial β-endorphin expression, separated from antineuroinflammation. Brain Behav Immun 2018; 73:504-519. [PMID: 29928964 DOI: 10.1016/j.bbi.2018.06.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/30/2018] [Accepted: 06/15/2018] [Indexed: 12/17/2022] Open
Abstract
Interleukin 10 (IL-10) is antinociceptive in various animal models of pain without induction of tolerance, and its mechanism of action was generally believed to be mediated by inhibition of neuroinflammation. Here we reported that intrathecal IL-10 injection dose dependently attenuated mechanical allodynia and thermal hyperalgesiain male and female neuropathic rats, with ED50 values of 40.8 ng and 24 ng, and Emax values of 61.5% MPE and 100% MPE in male rats. Treatment with IL-10 specifically increased expression of the β-endorphin (but not prodynorphin) gene and protein in primary cultures of spinal microglia but not in astrocytes or neurons. Intrathecal injection of IL-10 stimulated β-endorphin expression from microglia but not neurons or astrocytes in both contralateral and ipsilateral spinal cords of neuropathic rats. However, intrathecal injection of the β-endorphin neutralizing antibody, opioid receptor antagonist naloxone, or μ-opioid receptor antagonist CTAP completely blocked spinal IL-10-induced mechanical antiallodynia, while the microglial inhibitor minocycline and specific microglia depletor reversed spinal IL-10-induced β-endorphin overexpression and mechanical antiallodynia. IL-10 treatment increased spinal microglial STAT3 phosphorylation, and the STAT3 inhibitor NSC74859 completely reversed IL-10-increased spinal expression of β-endorphin and neuroinflammatory cytokines and mechanical antiallodynia. Silence of the Bcl3 and Socs3 genes nearly fully reversed IL-10-induced suppression of neuroinflammatory cytokines (but not expression of β-endorphin), although it had no effect on mechanical allodynia. In contrast, disruption of the POMC gene completely blocked IL-10-stimulated β-endorphin expression and mechanical antiallodynia, but had no effect on IL-10 inhibited expression of neuroinflammatory cytokines. Thus this study revealed that IL-10 produced antinociception through spinal microglial β-endorphin expression, but not inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Hai-Yun Wu
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Xiao-Fang Mao
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Xue-Qi Tang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Usman Ali
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Evhy Apryani
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Hao Liu
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Xin-Yan Li
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai 200240, China.
| |
Collapse
|
42
|
Prezotto LD, Thorson JF, Borowicz PP, Peine JL, Bedenbaugh M, Hileman SM, Lents CA, Caton JS, Swanson KC. Influences of maternal nutrient restriction and arginine supplementation on visceral metabolism and hypothalamic circuitry of offspring. Domest Anim Endocrinol 2018; 65:71-79. [PMID: 30007131 DOI: 10.1016/j.domaniend.2018.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/23/2018] [Accepted: 06/13/2018] [Indexed: 11/17/2022]
Abstract
Maternal nutrient restriction during gestation can exert long-term negative effects on offspring health and performance. Arginine supplementation may rescue some of the negative effects elicited by maternal nutrient restriction. We tested the hypothesis that maternal arginine supplementation during gestation would rescue deleterious effects of nutrient restriction on in vitro O2 consumption in the liver and jejunum and hypothalamic protein expression of proopiomelanocortin (POMC), neuropeptide Y (NPY), agouti-related peptide (AgRP), and neuronal nitric oxide synthase (nNOS), and the colocalization of nNOS and active phosphor-signal transducer and activator of transcription 3 (pSTAT3) in female offspring. Multiparous ewes were assigned to dietary treatment at 54 d of gestation: 100% of requirements (Con), 60% of control (Res), or Res plus rumen-protected arginine (Res-Arg; 180 mg/kg). At parturition, offspring were immediately removed from their dam and placed on a common diet. At 54 ± 4 d of age, female lambs (n = 6 per treatment) were weighed, the liver and jejunum were weighed, and samples were collected for in vitro measurement of O2 consumption. The hypothalamus was collected to determine protein expression of POMC, NPY, AgRP, and nNOS, and the colocalization of nNOS and pSTAT3 (n = 3, 4, and 4 for Con, Res, and Res-Arg, respectively). Hepatic consumption of O2 in vitro (mol/min/liver) was decreased (P = 0.04) in the Res and Res-Arg group compared with Con. Intensity of staining for NPY-containing fibers tended to decrease (P = 0.10) in Res and Res-Arg compared with Con. Number of POMC neuronal cells in the arcuate nucleus (ARC) of the hypothalamus decreased (P ≤ 0.03) in the Res group compared with Res-Arg. These observations demonstrate that maternal nutrient restriction decreases energy utilization in the liver and number of POMC cells in the ARC of offspring. Supplementation of arginine to the gestating ewe failed to influence hepatic use of energy in lambs from Res ewes. Numbers of POMC-containing cells were increased in the ARC in lambs from ewes restricted to 60% of nutritional requirements and supplemented with rumen-protected arginine, potentially influencing feeding behavior and hepatic energy metabolism.
Collapse
Affiliation(s)
- Ligia D Prezotto
- Department of Animal Sciences, North Dakota State University, Hultz Hall 166, Dept. 7630, PO Box 6050, Fargo 58108, USA; Northern Agricultural Research Center, Montana State University, 3710 Assinniboine Road, Havre 59501, USA
| | - Jennifer F Thorson
- Northern Agricultural Research Center, Montana State University, 3710 Assinniboine Road, Havre 59501, USA; USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center 68933, USA
| | - Pawel P Borowicz
- Department of Animal Sciences, North Dakota State University, Hultz Hall 166, Dept. 7630, PO Box 6050, Fargo 58108, USA
| | - Jena L Peine
- Department of Animal Sciences, North Dakota State University, Hultz Hall 166, Dept. 7630, PO Box 6050, Fargo 58108, USA
| | - Michelle Bedenbaugh
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Stanley M Hileman
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| | - Clay A Lents
- USDA, ARS, U.S. Meat Animal Research Center, P.O. Box 166, Clay Center 68933, USA
| | - Joel S Caton
- Department of Animal Sciences, North Dakota State University, Hultz Hall 166, Dept. 7630, PO Box 6050, Fargo 58108, USA
| | - Kendall C Swanson
- Department of Animal Sciences, North Dakota State University, Hultz Hall 166, Dept. 7630, PO Box 6050, Fargo 58108, USA.
| |
Collapse
|
43
|
Evely KM, Pryce KD, Bausch AE, Lukowski R, Ruth P, Haj-Dahmane S, Bhattacharjee A. Slack K Na Channels Influence Dorsal Horn Synapses and Nociceptive Behavior. Mol Pain 2018; 13:1744806917714342. [PMID: 28604221 PMCID: PMC5486487 DOI: 10.1177/1744806917714342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The sodium-activated potassium channel Slack (Kcnt1, Slo2.2) is highly expressed in dorsal root ganglion neurons where it regulates neuronal firing. Several studies have implicated the Slack channel in pain processing, but the precise mechanism or the levels within the sensory pathway where channels are involved remain unclear. Here, we furthered the behavioral characterization of Slack channel knockout mice and for the first time examined the role of Slack channels in the superficial, pain-processing lamina of the dorsal horn. We performed whole-cell recordings from spinal cord slices to examine the intrinsic and synaptic properties of putative inhibitory and excitatory lamina II interneurons. Slack channel deletion altered intrinsic properties and synaptic drive to favor an overall enhanced excitatory tone. We measured the amplitudes and paired pulse ratio of paired excitatory post-synaptic currents at primary afferent synapses evoked by electrical stimulation of the dorsal root entry zone. We found a substantial decrease in the paired pulse ratio at synapses in Slack deleted neurons compared to wildtype, indicating increased presynaptic release from primary afferents. Corroborating these data, plantar test showed Slack knockout mice have an enhanced nociceptive responsiveness to localized thermal stimuli compared to wildtype mice. Our findings suggest that Slack channels regulate synaptic transmission within the spinal cord dorsal horn and by doing so establishes the threshold for thermal nociception.
Collapse
Affiliation(s)
- Katherine M Evely
- Program in Neuroscience, University at Buffalo, The State University of New York, NY, USA
| | - Kerri D Pryce
- Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, NY, USA
| | - Anne E Bausch
- Institut für Pharmazie, Pharmakologie, Toxikologie und Klinische Pharmazie der Universität Tübingen, Germany
| | - Robert Lukowski
- Institut für Pharmazie, Pharmakologie, Toxikologie und Klinische Pharmazie der Universität Tübingen, Germany
| | - Peter Ruth
- Institut für Pharmazie, Pharmakologie, Toxikologie und Klinische Pharmazie der Universität Tübingen, Germany
| | - Samir Haj-Dahmane
- Program in Neuroscience, University at Buffalo, The State University of New York, NY, USA
| | | |
Collapse
|
44
|
Zhou Y, Kreek MJ. Involvement of Activated Brain Stress Responsive Systems in Excessive and "Relapse" Alcohol Drinking in Rodent Models: Implications for Therapeutics. J Pharmacol Exp Ther 2018; 366:9-20. [PMID: 29669731 PMCID: PMC5988024 DOI: 10.1124/jpet.117.245621] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/16/2018] [Indexed: 02/06/2023] Open
Abstract
Addictive diseases, including addiction to alcohol, pose massive public health costs. Addiction is a chronic relapsing disease caused by both the direct effects induced by drugs and persistent neuroadaptations at the molecular, cellular, and behavioral levels. These drug-type specific neuroadaptations are brought on largely by the reinforcing effects of drugs on the central nervous system and environmental stressors. Results from animal experiments have demonstrated important interactions between alcohol and stress-responsive systems. Addiction to specific drugs such as alcohol, psychostimulants, and opioids shares some common direct or downstream effects on the brain's stress-responsive systems, including arginine vasopressin and its V1b receptors, dynorphin and the κ-opioid receptors, pro-opiomelanocortin/β-endorphin and the μ-opioid receptors, and the endocannabinoids. Further study of these systems through laboratory-based and translational research could lead to the discovery of novel treatment targets and the early optimization of interventions (for example, combination) for the pharmacologic therapy of alcoholism.
Collapse
Affiliation(s)
- Yan Zhou
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Mary Jeanne Kreek
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, New York
| |
Collapse
|
45
|
Fregni F, Macedo I, Spezia-Adachi L, Scarabelot V, Laste G, Souza A, Sanches PRS, Caumo W, Torres I. Transcranial direct current stimulation (tDCS) prevents chronic stress-induced hyperalgesia in rats. Brain Stimul 2018; 11:299-301. [DOI: 10.1016/j.brs.2017.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022] Open
|
46
|
Weinsanto I, Mouheiche J, Laux-Biehlmann A, Aouad M, Maduna T, Petit-Demoulière N, Chavant V, Poisbeau P, Darbon P, Charlet A, Giersch A, Parat MO, Goumon Y. Lithium reverses mechanical allodynia through a mu opioid-dependent mechanism. Mol Pain 2018; 14:1744806917754142. [PMID: 29353538 PMCID: PMC5788089 DOI: 10.1177/1744806917754142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Lithium is widely used to treat bipolar disorders and displays mood stabilizing properties. In addition, lithium relieves painful cluster headaches and has a strong analgesic effect in neuropathic pain rat models. Objectives To investigate the analgesic effect of lithium on the cuff model of neuropathic pain. Methods We used behavioral and pharmacological approaches to study the analgesic effect of a single injection of lithium in wild-type and mu opioid receptor (MOR) null cuffed neuropathic mice. Mass spectrometry and enzyme-linked immunosorbent assay allowed to measure the levels of endogenous MOR agonist beta-endorphin as well as monoamines in brain and plasma samples 4 h after lithium administration. Results A single injection of lithium chloride (100 mg/kg, ip) alleviated mechanical allodynia for 24 h, and this effect was absent in MOR null neuropathic mice. Biochemical analyses highlight a significant increase in beta-endorphin levels by 30% in the brain of lithium-treated mice compared to controls. No variation of beta-endorphin was detected in the blood. Conclusions Together, our results provide evidence that lithium induces a long-lasting analgesia in neuropathic mice presumably through elevated brain levels of beta-endorphin and the activation of MORs.
Collapse
Affiliation(s)
- Ivan Weinsanto
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Jinane Mouheiche
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Alexis Laux-Biehlmann
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Maya Aouad
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Tando Maduna
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Nathalie Petit-Demoulière
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Virginie Chavant
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
- Mass Spectrometry Facilities, CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Pierrick Poisbeau
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Pascal Darbon
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Alexandre Charlet
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Anne Giersch
- INSERM U-1114, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Département de Psychiatrie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Marie-Odile Parat
- School of Pharmacy, University of Queensland, PACE, Woolloongabba, Australia
| | - Yannick Goumon
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- University of Strasbourg, Strasbourg, France
- Mass Spectrometry Facilities, CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
- Yannick Goumon, INCI, CNRS UPR3212, 5, rue Blaise Pascal, F-67084 Strasbourg Cedex, France.
| |
Collapse
|
47
|
Maldonado R, Baños JE, Cabañero D. Usefulness of knockout mice to clarify the role of the opioid system in chronic pain. Br J Pharmacol 2018; 175:2791-2808. [PMID: 29124744 DOI: 10.1111/bph.14088] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022] Open
Abstract
Several lines of knockout mice deficient in the genes encoding each component of the endogenous opioid system have been used for decades to clarify the specific role of the different opioid receptors and peptide precursors in many physiopathological conditions. The use of these genetically modified mice has improved our knowledge of the specific involvement of each endogenous opioid component in nociceptive transmission during acute and chronic pain conditions. The present review summarizes the recent advances obtained using these genetic tools in understanding the role of the opioid system in the pathophysiological mechanisms underlying chronic pain. Behavioural data obtained in these chronic pain models are discussed considering the peculiarities of the behavioural phenotype of each line of knockout mice. These studies have identified the crucial role of specific components of the opioid system in different manifestations of chronic pain and have also opened new possible therapeutic approaches, such as the development of opioid compounds simultaneously targeting several opioid receptors. However, several questions still remain open and require further experimental effort to be clarified. The novel genetic tools now available to manipulate specific neuronal populations and precise genome editing in mice will facilitate in a near future the elucidation of the role of each component of the endogenous opioid system in chronic pain. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Josep Eladi Baños
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
48
|
Pasternak GW. Mu Opioid Pharmacology: 40 Years to the Promised Land. ADVANCES IN PHARMACOLOGY 2018; 82:261-291. [DOI: 10.1016/bs.apha.2017.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
49
|
Ferdousi M, Finn DP. Stress-induced modulation of pain: Role of the endogenous opioid system. PROGRESS IN BRAIN RESEARCH 2018; 239:121-177. [DOI: 10.1016/bs.pbr.2018.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
50
|
Stojakovic A, Espinosa EP, Farhad OT, Lutfy K. Effects of nicotine on homeostatic and hedonic components of food intake. J Endocrinol 2017; 235:R13-R31. [PMID: 28814527 PMCID: PMC5578410 DOI: 10.1530/joe-17-0166] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 07/17/2017] [Indexed: 12/30/2022]
Abstract
Chronic tobacco use leads to nicotine addiction that is characterized by exaggerated urges to use the drug despite the accompanying negative health and socioeconomic burdens. Interestingly, nicotine users are found to be leaner than the general population. Review of the existing literature revealed that nicotine affects energy homeostasis and food consumption via altering the activity of neurons containing orexigenic and anorexigenic peptides in the brain. Hypothalamus is one of the critical brain areas that regulates energy balance via the action of these neuropeptides. The equilibrium between these two groups of peptides can be shifted by nicotine leading to decreased food intake and weight loss. The aim of this article is to review the existing literature on the effect of nicotine on food intake and energy homeostasis and report on the changes that nicotine brings about in the level of these peptides and their receptors that may explain changes in food intake and body weight induced by nicotine. Furthermore, we review the effect of nicotine on the hedonic aspect of food intake. Finally, we discuss the involvement of different subtypes of nicotinic acetylcholine receptors in the regulatory action of nicotine on food intake and energy homeostasis.
Collapse
Affiliation(s)
- Andrea Stojakovic
- Department of Pharmaceutical SciencesCollege of Pharmacy, Western University of Health Sciences, Pomona, California, USA
- Mitochondrial Neurobiology and Therapeutics LaboratoryMayo Clinic, Rochester, Minnesota, USA
| | - Enma P Espinosa
- Department of Pharmaceutical SciencesCollege of Pharmacy, Western University of Health Sciences, Pomona, California, USA
- Faculty of MedicineSchool of Clinica Biochemistry, Pontifical Catholic University of Ecuador (PUCE), Quito, Ecuador
| | - Osman T Farhad
- Department of Pharmaceutical SciencesCollege of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| | - Kabirullah Lutfy
- Department of Pharmaceutical SciencesCollege of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|