1
|
Quintanilla ME, Morales P, Santapau D, Gallardo J, Rebolledo R, Riveras G, Acuña T, Herrera-Marschitz M, Israel Y, Ezquer F. Morphine self-administration is inhibited by the antioxidant N-acetylcysteine and the anti-inflammatory ibudilast; an effect enhanced by their co-administration. PLoS One 2024; 19:e0312828. [PMID: 39471200 PMCID: PMC11521314 DOI: 10.1371/journal.pone.0312828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/11/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND The treatment of opioid addiction mainly involves the medical administration of methadone or other opioids, aimed at gradually reducing dependence and, consequently, the need for illicit opioid procurement. Thus, initiating opioid maintenance therapy with a lower level of dependence would be advantageous. There is compelling evidence indicating that opioids induce brain oxidative stress and associated glial activation, resulting in the dysregulation of glutamatergic homeostasis, which perpetuates drug intake. The present study aimed to determine whether inhibiting oxidative stress and/or neuroinflammation reduces morphine self-administration in an animal model of opioid dependence. METHODS Morphine dependence, assessed as voluntary morphine self-administration, was evaluated in Wistar-derived UChB rats. Following an extended period of morphine self-administration, animals were administered either the antioxidant N-acetylcysteine (NAC; 40 mg/kg/day), the anti-inflammatory ibudilast (7.5 mg/kg/day) or the combination of both agents. Oxidative stress and neuroinflammation were evaluated in the hippocampus, a region involved in drug recall that feeds into the nucleus accumbens, where the levels of the glutamate transporters GLT-1 and xCT were further assessed. RESULTS Daily administration of either NAC or ibudilast led to a mild reduction in voluntary morphine intake, while the co-administration of both therapeutic agents resulted in a marked inhibition (-57%) of morphine self-administration. The administration of NAC or ibudilast markedly reduced both the oxidative stress induced by chronic morphine intake and the activation of microglia and astrocytes in the hippocampus. However, only the combined administration of NAC + ibudilast was able to restore the normal levels of the glutamate transporter GLT-1 in the nucleus accumbens. CONCLUSION Separate or joint administration of an antioxidant and anti-inflammatory agent reduced voluntary opioid intake, which could have translational value for the treatment of opioid use disorders, particularly in settings where the continued maintenance of oral opioids is a therapeutic option.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for the Prevention of Substance use and the Treatment of Addictions (CESA), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for the Prevention of Substance use and the Treatment of Addictions (CESA), Faculty of Medicine, University of Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Daniela Santapau
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Rocío Rebolledo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Gabriel Riveras
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Tirso Acuña
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for the Prevention of Substance use and the Treatment of Addictions (CESA), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile
| |
Collapse
|
2
|
Williams IAR, Clemens KJ. Emerging evidence of a link between inflammation and the neuropathology of prenatal opioid exposure. Curr Opin Neurobiol 2024; 89:102924. [PMID: 39366149 DOI: 10.1016/j.conb.2024.102924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 10/06/2024]
Abstract
Opioid use continues to increase, particularly among women of reproductive age. As a result, increasing numbers of infants are born with prenatal exposure to opioids, suffering both acute and long-term negative consequences. Studies performed across the past 5 years have highlighted both peripheral and central inflammation as a consistent feature of prenatal opioid exposure. Dysregulated innate and adaptive immunity have been detected in human and rodent studies, highlighting a likely role of inflammation in the neuropathology associated with opioid exposure. Identifying immune changes occurring following prenatal opioid exposure will be critical for developing new therapeutic approaches in this field.
Collapse
Affiliation(s)
- Isobel A R Williams
- School of Psychology, University of New South Wales, Sydney, NSW 2052, Australia
| | - Kelly J Clemens
- School of Psychology, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
3
|
Hunter TJ, Videlefsky ZM, Ferreira Nakatani L, Zadina JE. Comparison of Morphine and Endomorphin Analog ZH853 for Tolerance and Immunomodulation in a Rat Model of Neuropathic Pain. THE JOURNAL OF PAIN 2024; 25:104607. [PMID: 38885918 DOI: 10.1016/j.jpain.2024.104607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
µ-Opioid receptor agonists, the gold standard for analgesia, come with significant side effects when used chronically. Tolerance, defined as the decrease in analgesic activity after repeated use, remains a vital therapeutic obstacle as it increases the likelihood of dose escalation and potentially lethal side effects like respiratory depression. Previous experiments have shown that the endomorphin-1 analog, ZH853, is a specific µ-opioid receptor agonist with reduced side effects like tolerance and glial activation following chronic central administration in pain-naive animals. Here, we investigated the effects of chronic, peripheral administration of µ-opioid receptor agonists following neuropathic injury. Though µ-opioids are effective at reducing neuropathic pain, they are not recommended for first-line treatment due to negative side effects. Compared with chronic morphine, chronic ZH853 treatment led to decreased tolerance and reduced glial activation. Following twice-daily intravenous injections, morphine was less potent and had a shorter duration of antinociception compared with ZH853. Chronic morphine, but not chronic ZH853, elevated markers of activation/inflammation of astrocytes (glial fibrillary acidic protein), microglia (ionized calcium-binding adapter molecule 1), the proinflammatory cytokine tumor necrosis factor-α, and phosphorylated mitogen-activated protein (MAP) kinase p38 (pp38). By contrast, chronic ZH853 reduced ionized calcium-binding adapter molecule 1 and tumor necrosis factor-α relative to both morphine and vehicle, suggesting anti-inflammatory properties with respect to these markers. Glial fibrillary acidic protein and pp38 were not significantly different from vehicle but were significantly lower than morphine. This study demonstrates the effectiveness of chronic ZH853 for providing analgesia in a neuropathic pain state with reduced tolerance compared with morphine, potentially due to reductions in spinal glial activation. PERSPECTIVE: Neuropathic pain is generally undertreated and resistant to medication, and side-effects limit opioid treatment. Here, we show that, compared with an equiantinociceptive dose of morphine, chronic intravenous administration of endomorphin analog ZH853 led to prolonged antiallodynia, reduced tolerance, and inhibition of spinal cord neuroinflammation in male spared nerve-injured rats.
Collapse
Affiliation(s)
- Terrence J Hunter
- Neuroscience Program/Brain Institute, Tulane University School of Medicine, New Orleans, Louisiana
| | - Zoe M Videlefsky
- Neuroscience Program/Brain Institute, Tulane University School of Medicine, New Orleans, Louisiana
| | | | - James E Zadina
- Neuroscience Program/Brain Institute, Tulane University School of Medicine, New Orleans, Louisiana; SE LA Veterans Health Care System, Tulane University School of Medicine, New Orleans, Louisiana; Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana; Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
4
|
Franklin JP, Testen A, Mieczkowski PA, Hepperla A, Crynen G, Simon JM, Wood JD, Harder EV, Bellinger TJ, Witt EA, Powell NL, Reissner KJ. Investigating cocaine- and abstinence-induced effects on astrocyte gene expression in the nucleus accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606656. [PMID: 39149305 PMCID: PMC11326167 DOI: 10.1101/2024.08.05.606656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
In recent years, astrocytes have been increasingly implicated in cellular mechanisms of substance use disorders (SUD). Astrocytes are structurally altered following exposure to drugs of abuse; specifically, astrocytes within the nucleus accumbens (NAc) exhibit significantly decreased surface area, volume, and synaptic colocalization after operant self-administration of cocaine and extinction or protracted abstinence (45 days). However, the mechanisms that elicit these morphological modifications are unknown. The current study aims to elucidate the molecular modifications that lead to observed astrocyte structural changes in rats across cocaine abstinence using astrocyte-specific RiboTag and RNA-seq, as an unbiased, comprehensive approach to identify genes whose transcription or translation change within NAc astrocytes following cocaine self-administration and extended abstinence. Using this method, our data reveal cellular processes including cholesterol biosynthesis that are altered specifically by cocaine self-administration and abstinence, suggesting that astrocyte involvement in these processes is changed in cocaine-abstinent rats. Overall, the results of this study provide insight into astrocyte functional adaptations that occur due to cocaine exposure or during cocaine withdrawal, which may pinpoint further mechanisms that contribute to cocaine-seeking behavior.
Collapse
Affiliation(s)
- Janay P Franklin
- Neuroscience Center, University of North Carolina at Chapel Hill
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Anze Testen
- Department of Neuroscience, Medical University of South Carolina
| | | | - Austin Hepperla
- Department of Genetics, University of North Carolina at Chapel Hill
| | - Gogce Crynen
- Bioinformatics and Statistics Core, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology
| | - Jeremy M Simon
- Department of Data Science, Dana-Farber Institute Department of Biostatistics, Harvard T.H. Chan School of Public Health
| | - Jonathan D Wood
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Eden V Harder
- Neuroscience Center, University of North Carolina at Chapel Hill
| | - Tania J Bellinger
- Department of Pharmacology, University of North Carolina at Chapel Hill
| | - Emily A Witt
- Department of Medical Neuroscience, Dalhousie University
| | - N LaShae Powell
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Kathryn J Reissner
- Neuroscience Center, University of North Carolina at Chapel Hill
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| |
Collapse
|
5
|
Truitt B, Tao J, Roy S. Impact of Morphine withdrawal on genes related to the TLR2 pathway expressed in the Prefrontal Cortex. Sci Prog 2024; 107:368504241264994. [PMID: 39228316 PMCID: PMC11375641 DOI: 10.1177/00368504241264994] [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: 09/05/2024]
Abstract
Millions of people suffer from opioid use disorder, because of the ongoing opioid epidemic. The aversive symptoms of withdrawal are a leading factor for drug relapses, yet there are limited therapeutic options to minimize or prevent withdrawal symptoms. The mechanism behind opioid withdrawal is still not fully understood, thus preventing the development of new therapeutics. This study is an extension of our previously proposed mechanism of a toll-like receptor 2 (TLR2) mediated withdrawal response as a result of morphine induced microbial change that occurs during morphine withdrawal. Transcriptome analysis of the pre-frontal cortex indicated that there was increased expression of genes related to TLR2 signaling in morphine withdrawal treated animals compared to placebo controls. Antibiotic treatment further altered TLR2 related genes, recovering some of the morphine induced effect and leading to additional suppression of some genes related to the TLR2 pathway. Morphine withdrawal induced gene expression was attenuated in a whole body TLR2 knockout model. These results provide more support that TLR2 plays an integral role in morphine withdrawal mechanisms and could be a potential therapeutic target to minimize opioid withdrawal associated co-morbidities.
Collapse
Affiliation(s)
- Bridget Truitt
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Junyi Tao
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sabita Roy
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
6
|
Parekh SV, Adams LO, Barkell GA, Paniccia JE, Reissner KJ, Lysle DT. Dorsal hippocampal astrocytes mediate the development of heroin withdrawal-enhanced fear learning. Psychopharmacology (Berl) 2024; 241:1265-1275. [PMID: 38396195 PMCID: PMC11106136 DOI: 10.1007/s00213-024-06562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
There is a significant co-occurrence of opioid use disorder (OUD) and post-traumatic stress disorder (PTSD) in clinical populations. However, the neurobiological mechanisms linking chronic opioid use, withdrawal, and the development of PTSD are poorly understood. Our previous research has shown that proinflammatory cytokines, expressed primarily by astrocytes in the dorsal hippocampus (DH), play a role in the development of heroin withdrawal-enhanced fear learning (HW-EFL), an animal model of PTSD-OUD comorbidity. Given the role of astrocytes in memory, fear learning, and opioid use, our experiments aimed to investigate their involvement in HW-EFL. Experiment 1 examined the effect of withdrawal from chronic heroin administration on GFAP surface area and volume, and identified increased surface area and volume of GFAP immunoreactivity in the dentate gyrus (DG) following 24-hour heroin withdrawal. Experiment 2 examined astrocyte morphology and synaptic interactions at the 24-hour withdrawal timepoint using an astroglial membrane-bound GFP (AAV5-GfaABC1D-lck-GFP). Although we did not detect significant changes in surface area and volume of GfaABC1D-Lck-GFP labelled astrocytes, we did observe a significant increase in the colocalization of astrocyte membranes with PSD-95 (postsynaptic density protein 95) in the DG. Experiment 3 tested if stimulating astroglial Gi signaling in the DH alters HW-EFL, and our results demonstrate this manipulation attenuates HW-EFL. Collectively, these findings contribute to our current understanding of the effects of heroin withdrawal on astrocytes and support the involvement of astrocytes in the comorbid relationship between opioid use and anxiety disorders.
Collapse
Affiliation(s)
- Shveta V Parekh
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Lydia O Adams
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Gillian A Barkell
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Jacqueline E Paniccia
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Kathryn J Reissner
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA
| | - Donald T Lysle
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, CB#3720, Chapel Hill, NC, 27599-3270, USA.
| |
Collapse
|
7
|
Bi K, Lei Y, Kong D, Li Y, Fan X, Luo X, Yang J, Wang G, Li X, Xu Y, Luo H. Progress in the study of intestinal microbiota involved in morphine tolerance. Heliyon 2024; 10:e27187. [PMID: 38533077 PMCID: PMC10963202 DOI: 10.1016/j.heliyon.2024.e27187] [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: 04/10/2023] [Revised: 01/09/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Morphine is a widely used opioid for treatment of pain. The attendant problems including morphine tolerance and morphine dependence pose a major public health challenge. In recent years, there has been increasing interest in the gastrointestinal microbiota in many physiological and pathophysiological processes. The connectivity network between the gut microbiota and the brain is involved in multiple biological systems, and bidirectional communication between them is critical in gastrointestinal tract homeostasis, the central nervous system, and the microbial system. Many research have previously shown that morphine has a variety of effects on the gastrointestinal tract, but none have determined the function of intestinal microbiota in morphine tolerance. This study reviewed the mechanisms of morphine tolerance from the perspective of dysregulation of microbiota-gut-brain axis homeostasis, by summarizing the possible mechanisms originating from the gut that may affect morphine tolerance and the improvement of morphine tolerance through the gut microbiota.
Collapse
Affiliation(s)
- Ke Bi
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Yi Lei
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Deshenyue Kong
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Yuansen Li
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Xuan Fan
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Xiao Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Jiqun Yang
- Third People's Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, 650041, China
| | - Guangqing Wang
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Xuejun Li
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Yu Xu
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Huayou Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
8
|
Wang B, Wang LN, Wu B, Guo R, Zhang L, Zhang JT, Wang ZH, Wu F, Feng Y, Liu H, Jin XH, Miao XH, Liu T. Astrocyte PERK and IRE1 Signaling Contributes to Morphine Tolerance and Hyperalgesia through Upregulation of Lipocalin-2 and NLRP3 Inflammasome in the Rodent Spinal Cord. Anesthesiology 2024; 140:558-577. [PMID: 38079113 DOI: 10.1097/aln.0000000000004858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
BACKGROUND Endoplasmic reticulum stress plays a crucial role in the pathogenesis of neuroinflammation and chronic pain. This study hypothesized that PRKR-like endoplasmic reticulum kinase (PERK) and inositol-requiring enzyme type 1 (IRE1) regulate lipocalin-2 (LCN2) and Nod-like receptor family pyrin domain containing 3 (NLRP3) expression in astrocytes, thereby contributing to morphine tolerance and hyperalgesia. METHODS The study was performed in Sprague-Dawley rats and C57/Bl6 mice of both sexes. The expression of LCN2 and NLRP3 was assessed by Western blotting. The tail-flick, von Frey, and Hargreaves tests were used to evaluate nociceptive behaviors. Chromatin immunoprecipitation was conducted to analyze the binding of activating transcription factor 4 (ATF4) to the promoters of LCN2 and TXNIP. Whole-cell patch-clamp recordings were used to evaluate neuronal excitability. RESULTS Pharmacologic inhibition of PERK and IRE1 attenuated the development of morphine tolerance and hyperalgesia in male (tail latency on day 7, 8.0 ± 1.13 s in the morphine + GSK2656157 [10 μg] group vs. 5.8 ± 0.65 s in the morphine group; P = 0.04; n = 6 rats/group) and female (tail latency on day 7, 6.0 ± 0.84 s in the morphine + GSK2656157 [10 μg] group vs. 3.1 ± 1.09 s in the morphine group; P = 0.0005; n = 6 rats/group) rats. Activation of PERK and IRE1 upregulated expression of LCN2 and NLRP3 in vivo and in vitro. Chromatin immunoprecipitation analysis showed that ATF4 directly bound to the promoters of the LCN2 and TXNIP. Lipocalin-2 induced neuronal hyperexcitability in the spinal cord and dorsal root ganglia via melanocortin-4 receptor. CONCLUSIONS Astrocyte endoplasmic reticulum stress sensors PERK and IRE1 facilitated morphine tolerance and hyperalgesia through upregulation of LCN2 and NLRP3 in the spinal cord. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Bing Wang
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China; Department of Pain Management, First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China; and Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey (current position)
| | - Li-Na Wang
- Department of Pain Management, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Wu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China
| | - Ran Guo
- Department of Pain, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Li Zhang
- Department of Anesthesiology, The First People's Hospital of Kunshan Affiliated with Jiangsu University, Kunshan, Jiangsu Province, China
| | - Jiang-Tao Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China
| | - Zhi-Hong Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Feng Wu
- Department of Pain Management, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Liu
- Department of Pain Management, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Hong Jin
- Department of Pain Management, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiu-Hua Miao
- Department of Pain, The Affiliated Hospital of Nantong University, Nantong, China
| | - Tong Liu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China; and College of Life Sciences, Yanan University, Yanan, China
| |
Collapse
|
9
|
Kupnicka P, Listos J, Tarnowski M, Kolasa A, Kapczuk P, Surówka A, Kwiatkowski J, Janawa K, Chlubek D, Baranowska-Bosiacka I. The Effect of Prenatal and Neonatal Fluoride Exposure to Morphine-Induced Neuroinflammation. Int J Mol Sci 2024; 25:826. [PMID: 38255899 PMCID: PMC10815549 DOI: 10.3390/ijms25020826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Physical dependence is associated with the formation of neuroadaptive changes in the central nervous system (CNS), both at the molecular and cellular levels. Various studies have demonstrated the immunomodulatory and proinflammatory properties of morphine. The resulting neuroinflammation in drug dependence exacerbates substance abuse-related behaviors and increases morphine tolerance. Studies prove that fluoride exposure may also contribute to the development of neuroinflammation and neurodegenerative changes. Morphine addiction is a major social problem. Neuroinflammation increases tolerance to morphine, and neurodegenerative effects caused by fluoride in structures related to the development of dependence may impair the functioning of neuronal pathways, change the concentration of neurotransmitters, and cause memory and learning disorders, which implies this element influences the development of dependence. Therefore, our study aimed to evaluate the inflammatory state of selected brain structures in morphine-dependent rats pre-exposed to fluoride, including changes in cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) expression as well as microglial and astroglial activity via the evaluation of Iba1 and GFAP expression. We provide evidence that both morphine administration and fluoride exposure have an impact on the inflammatory response by altering the expression of COX-1, COX-2, ionized calcium-binding adapter molecule (Iba1), and glial fibrillary acidic protein (GFAP) in brain structures involved in dependence development, such as the prefrontal cortex, striatum, hippocampus, and cerebellum. We observed that the expression of COX-1 and COX-2 in morphine-dependent rats is influenced by prior fluoride exposure, and these changes vary depending on the specific brain region. Additionally, we observed active astrogliosis, as indicated by increased GFAP expression, in all brain structures of morphine-dependent rats, regardless of fluoride exposure. Furthermore, the effect of morphine on Iba1 expression varied across different brain regions, and fluoride pre-exposure may influence microglial activation. However, it remains unclear whether these changes are a result of the direct or indirect actions of morphine and fluoride on the factors analyzed.
Collapse
Affiliation(s)
- Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Agnieszka Kolasa
- Department of Histology and Embryology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Anna Surówka
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Jakub Kwiatkowski
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Kamil Janawa
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
10
|
Harder EV, Franklin JP, VanRyzin JW, Reissner KJ. Astrocyte-Neuron Interactions in Substance Use Disorders. ADVANCES IN NEUROBIOLOGY 2024; 39:165-191. [PMID: 39190075 DOI: 10.1007/978-3-031-64839-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Engagement of astrocytes within the brain's reward circuitry has been apparent for approximately 30 years, when noncontingent drug administration was observed to lead to cytological markers of reactive astrocytes. Since that time, advanced approaches in rodent behavior and astrocyte monitoring have revealed complex interactions between astrocytes with drug type, animal sex, brain region, and dose and duration of drug administration. A number of studies now collectively reveal that rodent drug self-administration followed by prolonged abstinence results in decreased features of structure and synaptic colocalization of astrocytes. In addition, stimulation of astrocytes in the nucleus accumbens with DREADD receptors or pharmacological compounds opposes drug-seeking behavior. These findings provide a clear path for ongoing investigation into astrocytes as mediators of drug action in the brain and underscore the potential therapeutic utility of astrocytes in the regulation of drug craving and relapse vulnerability.
Collapse
Affiliation(s)
- Eden V Harder
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Janay P Franklin
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan W VanRyzin
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn J Reissner
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Quintanilla ME, Morales P, Santapau D, Ávila A, Ponce C, Berrios-Cárcamo P, Olivares B, Gallardo J, Ezquer M, Herrera-Marschitz M, Israel Y, Ezquer F. Chronic Voluntary Morphine Intake Is Associated with Changes in Brain Structures Involved in Drug Dependence in a Rat Model of Polydrug Use. Int J Mol Sci 2023; 24:17081. [PMID: 38069404 PMCID: PMC10707256 DOI: 10.3390/ijms242317081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Chronic opioid intake leads to several brain changes involved in the development of dependence, whereby an early hedonistic effect (liking) extends to the need to self-administer the drug (wanting), the latter being mostly a prefrontal-striatal function. The development of animal models for voluntary oral opioid intake represents an important tool for identifying the cellular and molecular alterations induced by chronic opioid use. Studies mainly in humans have shown that polydrug use and drug dependence are shared across various substances. We hypothesize that an animal bred for its alcohol preference would develop opioid dependence and further that this would be associated with the overt cortical abnormalities clinically described for opioid addicts. We show that Wistar-derived outbred UChB rats selected for their high alcohol preference additionally develop: (i) a preference for oral ingestion of morphine over water, resulting in morphine intake of 15 mg/kg/day; (ii) marked opioid dependence, as evidenced by the generation of strong withdrawal signs upon naloxone administration; (iii) prefrontal cortex alterations known to be associated with the loss of control over drug intake, namely, demyelination, axonal degeneration, and a reduction in glutamate transporter GLT-1 levels; and (iv) glial striatal neuroinflammation and brain oxidative stress, as previously reported for chronic alcohol and chronic nicotine use. These findings underline the relevance of polydrug animal models and their potential in the study of the wide spectrum of brain alterations induced by chronic morphine intake. This study should be valuable for future evaluations of therapeutic approaches for this devastating condition.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago 7610658, Chile; (M.E.Q.); (P.M.); (M.H.-M.); (Y.I.)
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago 7610658, Chile; (M.E.Q.); (P.M.); (M.H.-M.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago 7610658, Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 7610658, Chile
| | - Daniela Santapau
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile; (D.S.); (A.Á.); (P.B.-C.); (J.G.); (M.E.)
| | - Alba Ávila
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile; (D.S.); (A.Á.); (P.B.-C.); (J.G.); (M.E.)
| | - Carolina Ponce
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago 7610658, Chile
| | - Pablo Berrios-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile; (D.S.); (A.Á.); (P.B.-C.); (J.G.); (M.E.)
| | - Belén Olivares
- Center for Medical Chemistry, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile;
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile; (D.S.); (A.Á.); (P.B.-C.); (J.G.); (M.E.)
| | - Marcelo Ezquer
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile; (D.S.); (A.Á.); (P.B.-C.); (J.G.); (M.E.)
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago 7610658, Chile; (M.E.Q.); (P.M.); (M.H.-M.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago 7610658, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago 7610658, Chile; (M.E.Q.); (P.M.); (M.H.-M.); (Y.I.)
| | - Fernando Ezquer
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 7610658, Chile
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile; (D.S.); (A.Á.); (P.B.-C.); (J.G.); (M.E.)
| |
Collapse
|
12
|
Arai I, Tsuji M, Saito S, Takeda H. Experimental Study: Interleukin-31 Augments Morphine-Induced Antinociceptive Activity and Suppress Tolerance Development in Mice. Int J Mol Sci 2023; 24:16548. [PMID: 38003738 PMCID: PMC10671644 DOI: 10.3390/ijms242216548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Morphine-induced antinociception is partially reduced in interleukin-31 (IL-31) receptor A (IL-31RA)-deficient mice, indicating that IL-31RA is crucial for morphine-induced peripheral antinociception. Herein, we examined the combined effects of IL-31 and morphine on the antinociceptive activity and itch-associated scratching behavior (LLS) in mice and elucidated the regulatory mechanisms. A hot-plate test was used to assess antinociception. LLS was automatically detected and recorded via a computer. IL-31RA mRNA expression was assessed using real-time polymerase chain reaction. Repeated pre-treatment with IL-31 resulted in significant antinociceptive activity. Repeated administration of morphine decreased the morphine-induced antinociceptive activity, LLS counts, and regular dose and inhibited IL-31-induced LLS. These results suggested that the repeated administration of morphine depleted inter-neuronal IL-31RA levels, preventing morphine-induced antinociception. Therefore, IL-31 may be helpful as an adjunct analgesic to morphine. To explore the benefits of IL-31, its influence on morphine-induced antinociceptive tolerance in mice was examined. An IL-31 and morphine combination increased the analgesic action, which increased the expression of DRG neuronal IL-31RA, elucidating the site of peripheral antinociception of morphine. This site may induce exocytosis of IL-31RA in the sensory nervous system. Collectively, the suppressive effect of IL-31 on morphine-induced antinociceptive tolerance may result from IL-31RA supplementation in sensory nerves.
Collapse
Affiliation(s)
- Iwao Arai
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
- Division of Environmental Allergy, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Tokyo 105-8461, Japan
| | - Minoru Tsuji
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
| | - Saburo Saito
- Division of Environmental Allergy, The Jikei University School of Medicine, 3-25-8 Nishi-Shinbashi, Tokyo 105-8461, Japan
| | - Hiroshi Takeda
- Department of Pharmacology, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara 324-8510, Japan
| |
Collapse
|
13
|
Angeli A, Micheli L, Turnaturi R, Pasquinucci L, Parenti C, Alterio V, Di Fiore A, De Simone G, Monti SM, Carta F, Di Cesare Mannelli L, Ghelardini C, Supuran CT. Discovery of a novel series of potent carbonic anhydrase inhibitors with selective affinity for μ Opioid receptor for Safer and long-lasting analgesia. Eur J Med Chem 2023; 260:115783. [PMID: 37678143 DOI: 10.1016/j.ejmech.2023.115783] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
In this study, we investigated the development of dual-targeted ligands that bind to both μ-opioid receptor (MOR) and carbonic anhydrase (CA) enzymes, using fentanyl structure as a template. We synthesized and evaluated 21 novel compounds with dual-targeted affinity identifying the lead candidate compound 8, showing selective affinity for MOR and potent inhibition of several cytosolic CA isoforms. By means of repeated treatment of 3 daily administrations for 17 days, fentanyl (0.1 mg/kg, subcutaneously) led to tolerance development, pain threshold alterations and withdrawal symptoms in CD-1 mice, as well as astrocyte and microglia activation in the dorsal horn of the lumbar spinal cord. In contrast, compound 8 (0.32 mg/kg s.c.) maintained stable during days its analgesic effect at the higher dose tested with fewer withdrawal symptoms, allodynia development and glial cells activation. Our results suggest that targeting both MOR and CA enzymes can lead to the development of new class of potent analgesic agents with fewer side effects and reduced tolerance development. Further studies are needed to explore the potential mechanisms underlying these effects and to further optimize the therapeutic potential of these compounds.
Collapse
Affiliation(s)
- Andrea Angeli
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy.
| | - Laura Micheli
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini, 50139, Florence, Italy
| | - Rita Turnaturi
- Department of Drug Sciences and Health, Medicinal Chemistry Section, Università degli Studi di Catania, Viale A. Doria, 6, 95125, Catania, Italy
| | - Lorella Pasquinucci
- Department of Drug Sciences and Health, Medicinal Chemistry Section, Università degli Studi di Catania, Viale A. Doria, 6, 95125, Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences and Health, Pharmacology and Toxicology Section, Università degli Studi di Catania, Viale A. Doria, 6, 95125, Catania, Italy
| | - Vincenzo Alterio
- Istituto di Biostrutture e Bioimmagini-CNR, via Pietro Castellino, 111, 80131, Naples, Italy
| | - Anna Di Fiore
- Istituto di Biostrutture e Bioimmagini-CNR, via Pietro Castellino, 111, 80131, Naples, Italy
| | - Giuseppina De Simone
- Istituto di Biostrutture e Bioimmagini-CNR, via Pietro Castellino, 111, 80131, Naples, Italy
| | - Simona Maria Monti
- Istituto di Biostrutture e Bioimmagini-CNR, via Pietro Castellino, 111, 80131, Naples, Italy
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini, 50139, Florence, Italy
| | - Carla Ghelardini
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini, 50139, Florence, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
14
|
Phan TT, Jayathilake NJ, Lee KP, Park JM. BDNF/TrkB Signaling Inhibition Suppresses Astrogliosis and Alleviates Mechanical Allodynia in a Partial Crush Injury Model. Exp Neurobiol 2023; 32:343-353. [PMID: 37927132 PMCID: PMC10628862 DOI: 10.5607/en23031] [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: 09/26/2023] [Revised: 10/13/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023] Open
Abstract
Neuropathic pain presents a formidable clinical challenge due to its persistent nature and limited responsiveness to conventional analgesic treatments. While significant progress has been made in understanding the role of spinal astrocytes in neuropathic pain, their contribution and functional changes following a partial crush injury (PCI) remain unexplored. In this study, we investigated structural and functional changes in spinal astrocytes during chronic neuropathic pain, employing a partial crush injury model. This model allowes us to replicate the transition from initial nociceptive responses to persistent pain, highlighting the relevance of astrocytes in pain maintenance and sensitization. Through the examination of mechanical allodynia, a painful sensation in response to innocuous stimuli, and the correlation with increased levels of brain-derived neurotrophic factor (BDNF) along with reactive astrocytes, we identified a potential mechanistic link between astrocytic activity and BDNF signaling. Ultimately, our research provides evidence that inhibiting astrocyte activation through a BDNF/TrkB inhibitor alleviates mechanical allodynia, underscoring the therapeutic potential of targeting glial BDNF-related pathways for pain management. These findings offer critical insights into the cellular and molecular dynamics of neuropathic pain, paving the way for innovative and targeted treatment strategies for this challenging condition.
Collapse
Affiliation(s)
- Tien Thuy Phan
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
- IBS School, University of Science and Technology, Daejeon 34126, Korea
| | - Nishani Jayanika Jayathilake
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Kyu Pil Lee
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Joo Min Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
- IBS School, University of Science and Technology, Daejeon 34126, Korea
| |
Collapse
|
15
|
Suleymanova I, Bychkov D, Kopra J. A deep convolutional neural network for efficient microglia detection. Sci Rep 2023; 13:11139. [PMID: 37429956 PMCID: PMC10333175 DOI: 10.1038/s41598-023-37963-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/30/2023] [Indexed: 07/12/2023] Open
Abstract
Microglial cells are a type of glial cells that make up 10-15% of all brain cells, and they play a significant role in neurodegenerative disorders and cardiovascular diseases. Despite their vital role in these diseases, developing fully automated microglia counting methods from immunohistological images is challenging. Current image analysis methods are inefficient and lack accuracy in detecting microglia due to their morphological heterogeneity. This study presents development and validation of a fully automated and efficient microglia detection method using the YOLOv3 deep learning-based algorithm. We applied this method to analyse the number of microglia in different spinal cord and brain regions of rats exposed to opioid-induced hyperalgesia/tolerance. Our numerical tests showed that the proposed method outperforms existing computational and manual methods with high accuracy, achieving 94% precision, 91% recall, and 92% F1-score. Furthermore, our tool is freely available and adds value to exploring different disease models. Our findings demonstrate the effectiveness and efficiency of our new tool in automated microglia detection, providing a valuable asset for researchers in neuroscience.
Collapse
Affiliation(s)
- Ilida Suleymanova
- Faculty of Biological and Environmental Sciences, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
| | - Dmitrii Bychkov
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute for Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Amgott-Kwan AT, Zadina JE. Endomorphin analog ZH853 shows low reward, tolerance, and affective-motivational signs of withdrawal, while inhibiting opioid withdrawal and seeking. Neuropharmacology 2023; 227:109439. [PMID: 36709036 DOI: 10.1016/j.neuropharm.2023.109439] [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: 10/16/2022] [Revised: 01/09/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
Currently available μ-opioid receptor agonist pharmacotherapies for opioid use disorder possess adverse effects limiting their use and, despite treatment, rates of relapse remain high. We previously showed that endomorphin analog ZH853 had no effect in rodent models that predict abuse liability in humans. Here we extended these findings by examining dependence liability and reinforcing properties in female rats and male rats with previous opioid exposure. The potential use of ZH853 in managing opioid use disorder was evaluated by examining its effect on opioid-seeking behavior and withdrawal. We found that ZH853 did not induce locomotor activation in male and female mice and was not self-administered by female rats. Relative to morphine, ZH853 led to similar somatic signs of withdrawal, but low affective-motivational signs of withdrawal, and absent changes in ventral tegmental area K(+)-Cl(-) co-transporter expression associated with reward dysregulation. The low abuse liability of ZH853 was further supported in oxycodone self-administering male rats, where ZH853 substitution extinguished opioid-seeking behavior. ZH853 priming also did not reinstate morphine conditioned place preference. Lastly, ZH853 inhibited oxycodone-seeking behavior during relapse after forced abstinence and decreased the expression of morphine withdrawal. These findings suggest the potential use of ZH853 as a safer opioid medication for long-term treatment of pain and opioid use disorder.
Collapse
Affiliation(s)
- Ariel T Amgott-Kwan
- Neuroscience Program, Tulane Brain Institute: 6823 St Charles Avenue, 200 Flower Hall, Tulane University, New Orleans, LA, 70118, USA.
| | - James E Zadina
- Neuroscience Program, Tulane Brain Institute: 6823 St Charles Avenue, 200 Flower Hall, Tulane University, New Orleans, LA, 70118, USA; Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA; Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA; SE Louisiana Veterans Health Care System, 2400 Canal Street, New Orleans, LA, 70119, USA.
| |
Collapse
|
17
|
Ahmadi S, Mohammadi Talvar S, Masoudi K, Zobeiri M. Repeated Use of Morphine Induces Anxiety by Affecting a Proinflammatory Cytokine Signaling Pathway in the Prefrontal Cortex in Rats. Mol Neurobiol 2023; 60:1425-1439. [PMID: 36450935 DOI: 10.1007/s12035-022-03144-3] [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: 09/01/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022]
Abstract
We examined the role of toll-like receptors (TLRs) and proinflammatory cytokine signaling pathways in the prefrontal cortex (PFC) in anxiety-like behaviors after repeated use of morphine. Morphine (10 mg/kg) was used twice daily for 8 days to induce morphine dependence in male Wistar rats. On day 8, opioid dependence was confirmed by measuring naloxone-precipitated withdrawal signs. On days 1 and 8, anxiety-like behaviors were evaluated using a light/dark box test. Expression of TLR1 and 4, proinflammatory cytokines, and some of the downstream signaling molecules was also evaluated in the bilateral PFC at mRNA and protein levels following morphine dependence. The results revealed that morphine caused anxiolytic-like effects on day 1 while induced anxiety following 8 days of repeated injection. On day 8, a significant decrease in TLR1 expression was detected in the PFC in morphine-dependent rats, but TLR4 remained unaffected. Repeated morphine injection significantly increased IL1-β, TNFα, and IL6 expression, but decreased IL1R and TNFR at mRNA and protein levels except for IL6R at the protein level in the PFC. The p38α mitogen-activated protein (MAP) kinase expression significantly increased but the JNK3 expression decreased in the PFC in morphine-dependent rats. Repeated injection of morphine also significantly increased the NF-κB expression in the PFC. Further, significant increases in Let-7c, mir-133b, and mir-365 were detected in the PFC in morphine-dependent rats. We conclude that TLR1 and proinflammatory cytokines signaling pathways in the PFC are associated with the anxiogenic-like effects of morphine following its chronic use in rats via a MAP kinase/NF-κB pathway.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran.
| | - Shiva Mohammadi Talvar
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Kayvan Masoudi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Mohammad Zobeiri
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| |
Collapse
|
18
|
Cuitavi J, Torres-Pérez JV, Lorente JD, Campos-Jurado Y, Andrés-Herrera P, Polache A, Agustín-Pavón C, Hipólito L. Crosstalk between Mu-Opioid receptors and neuroinflammation: Consequences for drug addiction and pain. Neurosci Biobehav Rev 2023; 145:105011. [PMID: 36565942 DOI: 10.1016/j.neubiorev.2022.105011] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/29/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Mu-Opioid Receptors (MORs) are well-known for participating in analgesia, sedation, drug addiction, and other physiological functions. Although MORs have been related to neuroinflammation their biological mechanism remains unclear. It is suggested that MORs work alongside Toll-Like Receptors to enhance the release of pro-inflammatory mediators and cytokines during pathological conditions. Some cytokines, including TNF-α, IL-1β and IL-6, have been postulated to regulate MORs levels by both avoiding MOR recycling and enhancing its production. In addition, Neurokinin-1 Receptor, also affected during neuroinflammation, could be regulating MOR trafficking. Therefore, inflammation in the central nervous system seems to be associated with altered/increased MORs expression, which might regulate harmful processes, such as drug addiction and pain. Here, we provide a critical evaluation on MORs' role during neuroinflammation and its implication for these conditions. Understanding MORs' functioning, their regulation and implications on drug addiction and pain may help elucidate their potential therapeutic use against these pathological conditions and associated disorders.
Collapse
Affiliation(s)
- Javier Cuitavi
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain.
| | - Jose Vicente Torres-Pérez
- Department of Cellular Biology, Functional Biology and Physical Anthropology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Jesús David Lorente
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Yolanda Campos-Jurado
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Paula Andrés-Herrera
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Ana Polache
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Carmen Agustín-Pavón
- Department of Cellular Biology, Functional Biology and Physical Anthropology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain
| | - Lucía Hipólito
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicent Andrés Estellés s/n., 46100 Burjassot, Spain.
| |
Collapse
|
19
|
Liu X, Bae C, Liu B, Zhang YM, Zhou X, Zhang D, Zhou C, DiBua A, Schutz L, Kaczocha M, Puopolo M, Yamaguchi TP, Chung JM, Tang SJ. Development of opioid-induced hyperalgesia depends on reactive astrocytes controlled by Wnt5a signaling. Mol Psychiatry 2023; 28:767-779. [PMID: 36203006 PMCID: PMC10388343 DOI: 10.1038/s41380-022-01815-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/09/2022]
Abstract
Opioids are the frontline analgesics for managing various types of pain. Paradoxically, repeated use of opioid analgesics may cause an exacerbated pain state known as opioid-induced hyperalgesia (OIH), which significantly contributes to dose escalation and consequently opioid overdose. Neuronal malplasticity in pain circuits has been the predominant proposed mechanism of OIH expression. Although glial cells are known to become reactive in OIH animal models, their biological contribution to OIH remains to be defined and their activation mechanism remains to be elucidated. Here, we show that reactive astrocytes (a.k.a. astrogliosis) are critical for OIH development in both male and female mice. Genetic reduction of astrogliosis inhibited the expression of OIH and morphine-induced neural circuit polarization (NCP) in the spinal dorsal horn (SDH). We found that Wnt5a is a neuron-to-astrocyte signal that is required for morphine-induced astrogliosis. Conditional knock-out of Wnt5a in neurons or its co-receptor ROR2 in astrocytes blocked not only morphine-induced astrogliosis but also OIH and NCP. Furthermore, we showed that the Wnt5a-ROR2 signaling-dependent astrogliosis contributes to OIH via inflammasome-regulated IL-1β. Our results reveal an important role of morphine-induced astrogliosis in OIH pathogenesis and elucidate a neuron-to-astrocyte intercellular Wnt signaling pathway that controls the astrogliosis.
Collapse
Affiliation(s)
- Xin Liu
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Stony Brook University, Stony Brook, 11794, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Chilman Bae
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, 77555, TX, USA.,School of Electrical, Computer, and Biomedical Engineering, Southern Illinois University, Carbondale, 62901, IL, USA
| | - Bolong Liu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, 77555, TX, USA.,Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 W Tianhe Rd, Guangzhou, 510630, China
| | - Yong-Mei Zhang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, 77555, TX, USA.,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu Province, China
| | - Xiangfu Zhou
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 W Tianhe Rd, Guangzhou, 510630, China
| | - Donghang Zhang
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Adriana DiBua
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Stony Brook University, Stony Brook, 11794, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Livia Schutz
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Stony Brook University, Stony Brook, 11794, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Martin Kaczocha
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Stony Brook University, Stony Brook, 11794, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Michelino Puopolo
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Stony Brook University, Stony Brook, 11794, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 11794, NY, USA
| | - Terry P Yamaguchi
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, 21702, MD, USA
| | - Jin Mo Chung
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, 77555, TX, USA
| | - Shao-Jun Tang
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Stony Brook University, Stony Brook, 11794, NY, USA. .,Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, 11794, NY, USA. .,Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, 77555, TX, USA.
| |
Collapse
|
20
|
Toti A, Micheli L, Lucarini E, Ferrara V, Ciampi C, Margiotta F, Failli P, Gomiero C, Pallecchi M, Bartolucci G, Ghelardini C, Di Cesare Mannelli L. Ultramicronized N-Palmitoylethanolamine Regulates Mast Cell-Astrocyte Crosstalk: A New Potential Mechanism Underlying the Inhibition of Morphine Tolerance. Biomolecules 2023; 13:233. [PMID: 36830602 PMCID: PMC9953591 DOI: 10.3390/biom13020233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Persistent pain can be managed with opioids, but their use is limited by the onset of tolerance. Ultramicronized N-palmitoylethanolamine (PEA) in vivo delays morphine tolerance with mechanisms that are still unclear. Since glial cells are involved in opioid tolerance and mast cells (MCs) are pivotal targets of PEA, we hypothesized that a potential mechanism by which PEA delays opioid tolerance might depend on the control of the crosstalk between these cells. Morphine treatment (30 μM, 30 min) significantly increased MC degranulation of RBL-2H3 cells, which was prevented by pre-treatment with PEA (100 μM, 18 h), as evaluated by β-hexosaminidase assay and histamine quantification. The impact of RBL-2H3 secretome on glial cells was studied. Six-hour incubation of astrocytes with control RBL-2H3-conditioned medium, and even more so co-incubation with morphine, enhanced CCL2, IL-1β, IL-6, Serpina3n, EAAT2 and GFAP mRNA levels. The response was significantly prevented by the secretome from PEA pre-treated RBL-2H3, except for GFAP, which was further upregulated, suggesting a selective modulation of glial signaling. In conclusion, ultramicronized PEA down-modulated both morphine-induced MC degranulation and the expression of inflammatory and pain-related genes from astrocytes challenged with RBL-2H3 medium, suggesting that PEA may delay morphine tolerance, regulating MC-astrocyte crosstalk.
Collapse
Affiliation(s)
- Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Valentina Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Francesco Margiotta
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Paola Failli
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Chiara Gomiero
- Epitech Group SpA, Via Luigi Einaudi 13, 35030 Padua, Italy
| | - Marco Pallecchi
- Department of Chemistry, University of Florence, Via Ugo Schiff 6, 50019 Florence, Italy
| | - Gianluca Bartolucci
- Department of Chemistry, University of Florence, Via Ugo Schiff 6, 50019 Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| |
Collapse
|
21
|
El Jordi O, Fischer KD, Meyer TB, Atwood BK, Oblak AL, Pan RW, McKinzie DL. Microglial knockdown does not affect acute withdrawal but delays analgesic tolerance from oxycodone in male and female C57BL/6J mice. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10848. [PMID: 38390615 PMCID: PMC10880796 DOI: 10.3389/adar.2022.10848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/05/2022] [Indexed: 02/24/2024]
Abstract
Opioid Use Disorder (OUD) affects approximately 8%-12% of the population. In dependent individuals, abrupt cessation of opioid taking results in adverse withdrawal symptoms that reinforce drug taking behavior. Considerable unmet clinical need exists for new pharmacotherapies to treat opioid withdrawal as well as improve long-term abstinence. The neuroimmune system has received much scientific attention in recent years as a potential therapeutic target to combat various neurodegenerative and psychiatric disorders including addiction. However, the specific contribution of microglia has not been investigated in oxycodone dependence. Chronic daily treatment with the CSF1R inhibitor Pexidartinib (PLX3397) was administered to knockdown microglia expression and evaluate consequences on analgesia and on naloxone induced withdrawal from oxycodone. In vivo results indicated that an approximately 40% reduction in brain IBA1 staining was achieved in the PLX treatment group, which was associated with a delay in the development of analgesic tolerance to oxycodone and maintained antinociceptive efficacy. Acute withdrawal behavioral symptoms, brain astrocyte expression, and levels of many neuroinflammatory markers were not affected by PLX treatment. KC/GRO (also known as CXCL1) was significantly enhanced in the somatosensory cortex in oxycodone-treated mice receiving PLX. Microglial knock-down did not affect the expression of naloxoneinduced opioid withdrawal but affected antinociceptive responsivity. The consequences of increased KC/GRO expression within the somatosensory cortex due to microglial reduction during opioid dependence are unclear but may be important for neural pathways mediating opioid-induced analgesia.
Collapse
Affiliation(s)
- Omar El Jordi
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, United States
| | - Kathryn D Fischer
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, United States
| | - Timothy B Meyer
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, United States
| | - Brady K Atwood
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, United States
| | - Adrian L Oblak
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, United States
| | - Raymond W Pan
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, United States
| | - David L McKinzie
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
22
|
Effect of human mesenchymal stem cell secretome administration on morphine self-administration and relapse in two animal models of opioid dependence. Transl Psychiatry 2022; 12:462. [PMID: 36333316 PMCID: PMC9636200 DOI: 10.1038/s41398-022-02225-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
The present study investigates the possible therapeutic effects of human mesenchymal stem cell-derived secretome on morphine dependence and relapse. This was studied in a new model of chronic voluntary morphine intake in Wistar rats which shows classic signs of morphine intoxication and a severe naloxone-induced withdrawal syndrome. A single intranasal-systemic administration of MSCs secretome fully inhibited (>95%; p < 0.001) voluntary morphine intake and reduced the post-deprivation relapse intake by 50% (p < 0.02). Since several studies suggest a significant genetic contribution to the chronic use of many addictive drugs, the effect of MSCs secretome on morphine self-administration was further studied in rats bred as high alcohol consumers (UChB rats). Sub-chronic intraperitoneal administration of morphine before access to increasing concentrations of morphine solutions and water were available to the animals, led UChB rats to prefer ingesting morphine solutions over water, attaining levels of oral morphine intake in the range of those in the Wistar model. Intranasally administered MSCs secretome to UChB rats dose-dependently inhibited morphine self-administration by 72% (p < 0.001); while a single intranasal dose of MSC-secretome administered during a morphine deprivation period imposed on chronic morphine consumer UChB rats inhibited re-access morphine relapse intake by 80 to 85% (p < 0.0001). Both in the Wistar and the UChB rat models, MSCs-secretome administration reversed the morphine-induced increases in brain oxidative stress and neuroinflammation, considered as key engines perpetuating drug relapse. Overall, present preclinical studies suggest that products secreted by human mesenchymal stem cells may be of value in the treatment of opioid addiction.
Collapse
|
23
|
Ohashi Y, Sakhri FZ, Ikemoto H, Okumo T, Adachi N, Sunagawa M. Yokukansan Inhibits the Development of Morphine Tolerance by Regulating Presynaptic Proteins in DRG Neurons. Front Pharmacol 2022; 13:862539. [PMID: 35662731 PMCID: PMC9157577 DOI: 10.3389/fphar.2022.862539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Opioids, such as morphine, are used in clinical settings for the management of acute and chronic pain. However, long-term use of morphine leads to antinociceptive tolerance and hypersensitivity. The cellular and molecular mechanisms of morphine tolerance seem to be quite complex, with suggestions including internalization of the μ-opioid receptor (MOR), neuroinflammation with activation of microglia and astrocytes, and changes in synaptic function in the central nervous system. Yokukansan (YKS), a traditional Kampo medicine consisting of seven herbs, has been used to treat emotional instability, neurosis, and insomnia. Interestingly, recent studies have begun to reveal the inhibitory effect of YKS on the development of morphine tolerance. In the present study, we determined the effect of YKS on morphine tolerance formation and its mechanisms in a rat model, focusing on the synapses between primary sensory neurons and spinal dorsal horn secondary neurons. We found that morphine tolerance formation was significantly inhibited by YKS (0.3 or 1.0 g/kg/day) preadministration for 7 days. Repeated administration of morphine (10 mg/kg/day) increased the expression of presynaptic proteins, including synaptotagmin I, in the spinal cord, which was suppressed by YKS. Furthermore, these changes in presynaptic protein expression were more pronounced at isolectin B4 (IB4)-positive excitatory synapses around the lamina II of the dorsal horn. These results suggest that YKS suppresses the development of morphine tolerance by inhibiting the enhancement of presynaptic function of dorsal root ganglia neurons projecting to spinal dorsal horn neurons caused by continuous morphine administration.
Collapse
Affiliation(s)
- Yusuke Ohashi
- Department of Physiology, School of Medicine, Showa University, Tokyo, Japan
| | - Fatma Zahra Sakhri
- Department of Physiology, School of Medicine, Showa University, Tokyo, Japan
| | - Hideshi Ikemoto
- Department of Physiology, School of Medicine, Showa University, Tokyo, Japan
| | - Takayuki Okumo
- Department of Physiology, School of Medicine, Showa University, Tokyo, Japan
| | - Naoki Adachi
- Department of Physiology, School of Medicine, Showa University, Tokyo, Japan
| | - Masataka Sunagawa
- Department of Physiology, School of Medicine, Showa University, Tokyo, Japan
| |
Collapse
|
24
|
Hippocampus-sensitive and striatum-sensitive learning one month after morphine or cocaine exposure in male rats. Pharmacol Biochem Behav 2022; 217:173392. [PMID: 35513118 PMCID: PMC9796089 DOI: 10.1016/j.pbb.2022.173392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 12/31/2022]
Abstract
These experiments examined whether morphine and cocaine alter the balance between hippocampal and striatal memory systems measured long after drug exposure. Male rats received injections of morphine (5 mg/kg), cocaine (20 mg/kg), or saline for five consecutive days. One month later, rats were trained to find food on a hippocampus-sensitive place task or a striatum-sensitive response task. Relative to saline controls, morphine-treated rats exhibited impaired place learning but enhanced response learning; prior cocaine exposure did not significantly alter learning on either task. Another set of rats was trained on a dual-solution T-maze that can be solved with either place or response strategies. While a majority (67%) of control rats used place solutions, morphine treatment one month prior resulted in the exclusive use of response solutions (100%). Prior cocaine treatment did not significantly alter strategy selection. Molecular markers related to learning and drug abuse were measured in the hippocampus and striatum one month after drug exposure in behaviorally untested rats. Protein levels of glial-fibrillary acidic protein (GFAP), an intermediate filament specific to astrocytes, increased significantly in the hippocampus after morphine exposure, but not after cocaine exposure. Exposure to morphine or cocaine did not significantly change levels of brain-derived neurotrophic factor (BDNF) or a downstream target of BDNF signaling, glycogen synthase kinase 3β (GSK3β), in the hippocampus or striatum. Thus, exposure to morphine resulted in a long-lasting shift from hippocampal toward striatal dominance during learning, an effect that may be associated with lasting alterations in hippocampal astrocytes. Cocaine produced changes in the same direction, suggesting that use of a higher dose or longer duration of exposure might produce effects comparable to those seen with morphine.
Collapse
|
25
|
He L, Xu W, Zhang C, Ding Z, Guo Q, Zou W, Wang J. Dysregulation of Vesicular Glutamate Transporter VGluT2 via BDNF/TrkB Pathway Contributes to Morphine Tolerance in Mice. Front Pharmacol 2022; 13:861786. [PMID: 35559256 PMCID: PMC9086316 DOI: 10.3389/fphar.2022.861786] [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: 01/25/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
Morphine is widely used in the treatment of moderate to severe pain. Long-term use of morphine leads to various adverse effects, such as tolerance and hyperalgesia. Vesicular glutamate transporter 2 (VGluT2) accumulates glutamate into synaptic vesicles and plays multiple roles in the central nervous system. However, the specific role of VGluT2 in morphine tolerance has not been fully elucidated. Here, we investigated the regulatory role of VGluT2 in morphine tolerance and assessed the potential role of the brain-derived neurotrophic factor (BDNF)/tyrosine kinase B (TrkB) pathway in VGluT2 mediated morphine antinociceptive tolerance in mice. In the present study, we found that VGluT2 is upregulated in the spinal cord after the development of morphine tolerance. Furthermore, inhibition of VGluT2 with its antagonist (Chicago sky blue 6 B, CSB6B) or knockdown of VGluT2 by lentivirus restored the analgesic effect of morphine, suppressed the activation of astrocytes and microglia, and decreased glial-derived pro-inflammatory cytokines. Overexpression of VGluT2 by lentivirus facilitated morphine tolerance and mechanical hyperalgesia. In addition, we found the expression of BDNF is correlated with VGluT2 expression in the spinal cord after chronic morphine administration. Intrathecal injection of the BDNF/TrkB pathway antagonist K252a attenuated the development of morphine tolerance and decreased the expression of VGluT2 in the spinal cord, which suggested the BDNF/TrkB pathway participates in the regulation of VGluT2 in morphine tolerance. This study elucidates the functional capability of VGluT2 in modulating morphine tolerance and identifies a novel mechanism and promising therapeutic target for morphine tolerance.
Collapse
Affiliation(s)
- Liqiong He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xu
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuofeng Ding
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Thomas JHL, Lui L, Abell A, Tieu W, Somogyi AA, Bajic JE, Hutchinson MR. Toll-like receptors change morphine-induced antinociception, tolerance and dependence: Studies using male and female TLR and signalling gene KO mice. Brain Behav Immun 2022; 102:71-85. [PMID: 35131445 DOI: 10.1016/j.bbi.2022.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/22/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptors (TLR) have been proposed as a site of action that alters opioid pharmacodynamics. However, a comprehensive assessment of acute opioid antinociception, tolerance and withdrawal behaviours in genetic null mutant strains with altered innate immune signalling has not been performed. Nor has the impact of genetic deletion of TLR2/4 on high-affinity opioid receptor binding. Here we show that diminished TLR signalling potentiates acute morphine antinociception equally in male and female mice. However, only male TIR8 null mutant mice showed reduced morphine analgesia. Analgesic tolerance was prevented in TLR2 and TLR4 null mutants, but not MyD88 animals. Withdrawal behaviours were only protected in TLR2-/- mice. In silico docking simulations revealed opioid ligands bound preferentially to the LPS binding pocket of MD-2 rather than TLR4. There was no binding of [3H](-)-naloxone or [3H]diprenorphine to TLR4 in the concentrations explored. These data confirm that opioids have high efficacy activity at innate immune pattern recognition binding sites but do not bind to TLR4 and identify critical pathway and sex-specific effects of the complex innate immune signalling contributions to opioid pharmacodynamics. These data further support the behavioural importance of the TLR-opioid interaction but fail to demonstrate direct evidence for high-affinity binding of the TLR4 signalling complex to ligands.
Collapse
Affiliation(s)
- Jacob H L Thomas
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Liang Lui
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew Abell
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; ARC Centre for Nanoscale BioPhotonics, University of Adelaide, SA 5005, Australia
| | - William Tieu
- Discipline of Chemistry, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew A Somogyi
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Juliana E Bajic
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; ARC Centre for Nanoscale BioPhotonics, University of Adelaide, SA 5005, Australia
| | - Mark R Hutchinson
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; ARC Centre for Nanoscale BioPhotonics, University of Adelaide, SA 5005, Australia.
| |
Collapse
|
27
|
Lai H, Mubashir T, Shiwalkar N, Ahmad H, Balogh J, Williams G, Bauer C, Maroufy V. Association of pre-admission opioid abuse and/or dependence on major complications in traumatic brain injury (TBI) patients. J Clin Anesth 2022; 79:110719. [PMID: 35276593 DOI: 10.1016/j.jclinane.2022.110719] [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: 09/13/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 11/17/2022]
Abstract
SETTING In the last few decades, an opioid related health crisis has been a challenging problem in many countries around the world, especially the United States. Better understanding of the association of pre-admission opioid abuse and/or dependence (POAD) on specific major complications in traumatic brain injury (TBI) patients can aid the medical team in improving patient care management and outcomes. STUDY OBJECTIVE Our goal is to assess and quantify the risk of POAD on in-hospital mortality and major complications in TBI patients. DESIGN We conducted a retrospective study and used the National Inpatient Sample (NIS) database from 2004 to 2015 to investigate the impact of POAD on in-hospital mortality and major complications in TBI patients. We utilized propensity score matching and conditional logistic regression models, adjusted with injury severity score (ISS) and comorbidities, to obtain the adjusted odds ratios (OR). MAIN RESULTS POAD TBI patients had lower risks of in-hospital mortality (OR 0.58; p < 0.001) and acute myocardial infarction (OR 0.53; p = 0.045), while a higher risk of respiratory (OR 1.59; p < 0.001) and neurologic complications (OR 2.54; p < 0.001), compared to non-POAD TBI patients. Additionally, POAD patients were significantly more likely to have sepsis (OR 2.16, p < 0.001), malnutrition (OR 1.56, p < 0.001), delirium (OR 2.81, p < 0.001), respiratory failure (OR 1.79, p < 0.001), and acute renal failure (OR 1.83, p < 0.001). POAD TBI patients had shorter length of hospital stay compared to non-POAD TBI patients (mean 8.0 vs 9.2 days, p < 0.001). CONCLUSIONS POAD TBI patients have a lower in-hospital mortality, shorter duration of hospitalization and a lower risk of acute myocardial infarction, while they are more likely to have respiratory failure, delirium, sepsis, malnutrition, and acute renal failure compared to TBI patients without POAD. Prospective study is warranted to further confirm these findings.
Collapse
Affiliation(s)
- Hongyin Lai
- Department of Biostatistics and Data Science, UTHealth School of Public Health, 1200 Pressler St, Houston, TX 77030, USA
| | - Talha Mubashir
- Department of Anesthesiology, UTHealth McGovern Medical School, 6410 Fannin St, Houston, TX 77030, USA
| | - Nimisha Shiwalkar
- Department of Anesthesiology and Critical Care, Zen Multispecialty Hospital and Research Center, Mumbai, India
| | - Hunza Ahmad
- Department of Anesthesiology, UTHealth McGovern Medical School, 6410 Fannin St, Houston, TX 77030, USA
| | - Julius Balogh
- Department of Anesthesiology, University of Arkansas for Medical Sciences, 4301 West Markham, #515, Little Rock, AR 72205, USA
| | - George Williams
- Department of Anesthesiology, UTHealth McGovern Medical School, 6410 Fannin St, Houston, TX 77030, USA
| | - Cici Bauer
- Department of Biostatistics and Data Science, UTHealth School of Public Health, 1200 Pressler St, Houston, TX 77030, USA
| | - Vahed Maroufy
- Department of Biostatistics and Data Science, UTHealth School of Public Health, 1200 Pressler St, Houston, TX 77030, USA.
| |
Collapse
|
28
|
Somogyi AA, Musolino ST, Barratt DT. New pharmacological perspectives and therapeutic options for opioids: Differences matter. Anaesth Intensive Care 2022; 50:127-140. [PMID: 35112584 DOI: 10.1177/0310057x211063891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Opioids remain the major drug class for the treatment of acute, chronic and cancer pain, but have major harmful effects such as dependence and opioid-induced ventilatory impairment. Although no new typical opioids have come onto the market in the past almost 50 years, a plethora of new innovative formulations has been developed to meet the clinical need. This review is intended to shed light on new understanding of the molecular pharmacology of opioids, which has arisen largely due to the genomic revolution, and what new drugs may become available in the coming years. Atypical opioids have and are being developed which not only target the mu opioid receptor but other targets in the pain pathway. Biased mu agonists have been developed but remain 'unbiased' clinically. The contribution of drugs targeting non-mu opioid receptors either alone or as heterodimers shows potential promise but remains understudied. That gene splice variants of the mu opioid receptor produce multiple receptor isoforms in different brain regions, and may change with pain chronicity and phenotype, presents new challenges but also opportunities for precision pain medicine. Finally, that opioids also have pro-inflammatory effects not aligned with mu opioid receptor binding affinity implicates a fresh understanding of their role in chronic pain, whether cancer or non-cancer. Hopefully, a new understanding of opioid analgesic drug action may lead to new drug development and better precision medicine in acute and chronic pain relief with less patient harm.
Collapse
Affiliation(s)
- Andrew A Somogyi
- Discipline of Pharmacology, University of Adelaide, Adelaide, Australia
| | - Stefan T Musolino
- Discipline of Pharmacology, University of Adelaide, Adelaide, Australia
| | - Daniel T Barratt
- Discipline of Physiology, University of Adelaide, Adelaide, Australia
| |
Collapse
|
29
|
Düzenli N, Ülker S, Şengül G, Kayhan B, Önal A. Effects of cyanocobalamin and its combination with morphine on neuropathic rats and the relationship between these effects and thrombospondin-4 expression. Korean J Pain 2022; 35:66-77. [PMID: 34966013 PMCID: PMC8728557 DOI: 10.3344/kjp.2022.35.1.66] [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: 09/16/2021] [Revised: 12/04/2021] [Accepted: 12/04/2021] [Indexed: 11/06/2022] Open
Abstract
Background Thrombospondin-4 (TSP4) upregulates in the spinal cord following peripheral nerve injury and contributes to the development of neuropathic pain (NP). We investigated the effects of cyanocobalamin alone or in combination with morphine on pain and the relationship between these effects and spinal TSP4 expression in neuropathic rats. Methods NP was induced by chronic constriction injury (CCI) of the sciatic nerve. Cyanocobalamin (5 and 10 mg/kg/day) was administered 15 days before CCI and then for 4 and 14 postoperative days. Morphine (2.5 and 5 mg/kg/day) was administered only post-CCI. Combination treatment included cyanocobalamin and morphine, 10 and 5 mg/kg/day, respectively. All drugs were administered intraperitoneally. Nociceptive thresholds were detected by esthesiometer, analgesia meter, and plantar test, and TSP4 expression was assessed by western blotting and fluorescence immunohistochemistry. Results CCI decreased nociceptive thresholds in all tests and induced TSP4 expression on the 4th postoperative day. The decrease in nociceptive thresholds persisted except for the plantar test, and the increased TSP4 expression reversed on the 14th postoperative day. Cyanocobalamin and low-dose morphine alone did not produce any antinociceptive effects. High-dose morphine improved the decreased nociceptive thresholds in the esthesiometer when administered alone but combined with cyanocobalamin in all tests. Cyanocobalamin and morphine significantly induced TSP4 expression when administered alone in both doses for 4 or 14 days. However, this increase was less when the two drugs are combined. Conclusions The combination of cyanocobalamin and morphine is more effective in antinociception and partially decreased the induced TSP4 expression compared to the use of either drug alone.
Collapse
Affiliation(s)
- Neslihan Düzenli
- Department of Medical Pharmacology, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Sibel Ülker
- Department of Medical Pharmacology, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Gülgün Şengül
- Department of Anatomy, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Buse Kayhan
- Department of Neuroscience, Faculty of Health Sciences, Ege University, Bornova, Izmir, Turkey
| | - Aytül Önal
- Department of Medical Pharmacology, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey
| |
Collapse
|
30
|
Pati D, Kash TL. Tumor necrosis factor-α modulates GABAergic and dopaminergic neurons in the ventrolateral periaqueductal gray of female mice. J Neurophysiol 2021; 126:2119-2129. [PMID: 34817244 PMCID: PMC8715045 DOI: 10.1152/jn.00251.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/22/2022] Open
Abstract
Neuroimmune signaling is increasingly identified as a critical component of various illnesses, including chronic pain, substance use disorder, and depression. However, the underlying neural mechanisms remain unclear. Proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α), may play a role by modulating synaptic function and long-term plasticity. The midbrain structure periaqueductal gray (PAG) plays a well-established role in pain processing, and although TNF-α inhibitors have emerged as a therapeutic strategy for pain-related disorders, the impact of TNF-α on PAG neuronal activity has not been thoroughly characterized. Recent studies have identified subpopulations of ventrolateral PAG (vlPAG) with opposing effects on nociception, with dopamine (DA) neurons driving pain relief in contrast to GABA neurons. Therefore, we used slice physiology to examine the impact of TNF-α on neuronal activity of both these subpopulations. We focused on female mice since the PAG is a sexually dimorphic region and most studies use male subjects, limiting our understanding of mechanistic variations in females. We selectively targeted GABA and DA neurons using transgenic reporter lines. Following exposure to TNF-α, there was an increase in excitability of GABA neurons along with a reduction in glutamatergic synaptic transmission. In DA neurons, TNF-α exposure resulted in a robust decrease in excitability along with a modest reduction in glutamatergic synaptic transmission. Interestingly, TNF-α had no effect on inhibitory transmission onto DA neurons. Collectively, these data suggest that TNF-α differentially affects the function of GABA and DA neurons in female mice and enhances our understanding of how TNF-α-mediated signaling modulates vlPAG function.NEW & NOTEWORTHY This study describes the effects of TNF-α on two distinct subpopulations of neurons in the vlPAG. We show that TNF-α alters both neuronal excitability and glutamatergic synaptic transmission on GABA and dopamine neurons within the vlPAG of female mice. This provides critical new information on the role of TNF-α in the potential modulation of pain, since activation of vlPAG GABA neurons drives nociception, whereas activation of dopamine neurons drives analgesia.
Collapse
Affiliation(s)
- Dipanwita Pati
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
31
|
Pawlik K, Ciechanowska A, Ciapała K, Rojewska E, Makuch W, Mika J. Blockade of CC Chemokine Receptor Type 3 Diminishes Pain and Enhances Opioid Analgesic Potency in a Model of Neuropathic Pain. Front Immunol 2021; 12:781310. [PMID: 34795678 PMCID: PMC8593225 DOI: 10.3389/fimmu.2021.781310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 11/26/2022] Open
Abstract
Neuropathic pain is a serious clinical issue, and its treatment remains a challenge in contemporary medicine. Thus, dynamic development in the area of animal and clinical studies has been observed. The mechanisms of neuropathic pain are still not fully understood; therefore, studies investigating these mechanisms are extremely important. However, much evidence indicates that changes in the activation and infiltration of immune cells cause the release of pronociceptive cytokines and contribute to neuropathic pain development and maintenance. Moreover, these changes are associated with low efficacy of opioids used to treat neuropathy. To date, the role of CC chemokine receptor type 3 (CCR3) in nociception has not been studied. Similarly, little is known about its endogenous ligands (C-C motif ligand; CCL), namely, CCL5, CCL7, CCL11, CCL24, CCL26, and CCL28. Our research showed that the development of hypersensitivity in rats following chronic constriction injury (CCI) of the sciatic nerve is associated with upregulation of CCL7 and CCL11 in the spinal cord and dorsal root ganglia (DRG). Moreover, our results provide the first evidence that single and repeated intrathecal administration of the CCR3 antagonist SB328437 diminishes mechanical and thermal hypersensitivity. Additionally, repeated administration enhances the analgesic properties of morphine and buprenorphine following nerve injury. Simultaneously, the injection of SB328437 reduces the protein levels of some pronociceptive cytokines, such as IL-6, CCL7, and CCL11, in parallel with a reduction in the activation and influx of GFAP-, CD4- and MPO-positive cells in the spinal cord and/or DRG. Moreover, we have shown for the first time that an inhibitor of myeloperoxidase-4-aminobenzoic hydrazide may relieve pain and simultaneously enhance morphine and buprenorphine efficacy. The obtained results indicate the important role of CCR3 and its modulation in neuropathic pain treatment and suggest that it represents an interesting target for future investigations.
Collapse
Affiliation(s)
- Katarzyna Pawlik
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Agata Ciechanowska
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Ciapała
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewelina Rojewska
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
32
|
Allen MC, Moog NK, Buss C, Yen E, Gustafsson HC, Sullivan EL, Graham AM. Co-occurrence of preconception maternal childhood adversity and opioid use during pregnancy: Implications for offspring brain development. Neurotoxicol Teratol 2021; 88:107033. [PMID: 34601061 PMCID: PMC8578395 DOI: 10.1016/j.ntt.2021.107033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022]
Abstract
Understanding of the effects of in utero opioid exposure on neurodevelopment is a priority given the recent dramatic increase in opioid use among pregnant individuals. However, opioid abuse does not occur in isolation-pregnant individuals abusing opioids often have a significant history of adverse experiences in childhood, among other co-occurring factors. Understanding the specific pathways in which these frequently co-occurring factors may interact and cumulatively influence offspring brain development in utero represents a priority for future research in this area. We highlight maternal history of childhood adversity (CA) as one such co-occurring factor that is more prevalent among individuals using opioids during pregnancy and which is increasingly shown to affect offspring neurodevelopment through mechanisms beginning in utero. Despite the high incidence of CA history in pregnant individuals using opioids, we understand very little about the effects of comorbid prenatal opioid exposure and maternal CA history on fetal brain development. Here, we first provide an overview of current knowledge regarding effects of opioid exposure and maternal CA on offspring neurodevelopment that may occur during gestation. We then outline potential mechanistic pathways through which these factors might have interactive and cumulative influences on offspring neurodevelopment as a foundation for future research in this area.
Collapse
Affiliation(s)
- Madeleine C Allen
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Nora K Moog
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany; Development, Health and Disease Research Program, University of California, Irvine, 837 Health Sciences Drive, Irvine, California 92697, United States
| | - Elizabeth Yen
- Department of Pediatrics, Tufts Medical Center, Boston, MA 02111, United States
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185(th) Ave., Beaverton, OR 97006, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Alice M Graham
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States.
| |
Collapse
|
33
|
Interaction of Opioids with TLR4-Mechanisms and Ramifications. Cancers (Basel) 2021; 13:cancers13215274. [PMID: 34771442 PMCID: PMC8582379 DOI: 10.3390/cancers13215274] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/09/2021] [Accepted: 10/17/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Recent evidence indicates that opioids can be active at a receptor that is abundantly expressed on innate immune cells as well as cancer cells: the receptor is termed toll-like receptor 4 (TLR4). TLR4 is increasingly recognised as playing key roles in tumour biology and anticancer defences. However, the issue of whether TLR4 mediates some of the effects of opioids on tumour growth and metastasis is entirely unknown. We review existing evidence, mechanisms, and functional consequences of the action of opioids at TLR4. This opens new avenues of research on the role of opioids in cancer. Abstract The innate immune receptor toll-like receptor 4 (TLR4) is known as a sensor for the gram-negative bacterial cell wall component lipopolysaccharide (LPS). TLR4 activation leads to a strong pro-inflammatory response in macrophages; however, it is also recognised to play a key role in cancer. Recent studies of the opioid receptor (OR)-independent actions of opioids have identified that TLR4 can respond to opioids. Opioids are reported to weakly activate TLR4, but to significantly inhibit LPS-induced TLR4 activation. The action of opioids at TLR4 is suggested to be non-stereoselective, this is because OR-inactive (+)-isomers of opioids have been shown to activate or to inhibit TLR4 signalling, although there is some controversy in the literature. While some opioids can bind to the lipopolysaccharide (LPS)-binding cleft of the Myeloid Differentiation factor 2 (MD-2) co-receptor, pharmacological characterisation of the inhibition of opioids on LPS activation of TLR4 indicates a noncompetitive mechanism. In addition to a direct interaction at the receptor, opioids affect NF-κB activation downstream of both TLR4 and opioid receptors and modulate TLR4 expression, leading to a range of in vivo outcomes. Here, we review the literature reporting the activity of opioids at TLR4, its proposed mechanism(s), and the complex functional consequences of this interaction.
Collapse
|
34
|
Astrocyte-Derived Extracellular Vesicle-Mediated Activation of Primary Ciliary Signaling Contributes to the Development of Morphine Tolerance. Biol Psychiatry 2021; 90:575-585. [PMID: 34417054 DOI: 10.1016/j.biopsych.2021.06.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Morphine is used extensively in the clinical setting owing to its beneficial effects, such as pain relief; its therapeutic utility is limited because the prolonged use of morphine often results in tolerance and addiction. Astrocytes in the brain are a direct target of morphine action and play an essential role in the development of morphine tolerance. Primary cilia and the cilia-mediated sonic hedgehog (SHH) signaling pathways have been shown to play a role in drug resistance and morphine tolerance, respectively. Extracellular vesicles (EVs) play important roles as cargo-carrying vesicles mediating communication among cells and tissues. METHODS C57BL/6N mice were administered morphine for 8 days to develop tolerance, which was determined using the tail-flick and hot plate assays. EVs were separated from astrocyte-conditioned media using either size exclusion chromatography or ultracentrifugation approaches, followed by characterization of EVs using nanoparticle tracking analysis for EV size distribution and number, Western blotting for EV markers, and electron microscopy for EV morphology. Astrocytes were treated with EVs for 24 hours, followed by assessing primary cilia by fluorescent immunostaining for primary cilia markers (ARL13B and acetylated tubulin). RESULTS Morphine-tolerant mice exhibited an increase in primary cilia length and percentage of ciliated astrocytes. The levels of SHH protein were upregulated in morphine-stimulated astrocyte-derived EVs. SHH on morphine-stimulated astrocyte-derived EVs activated SHH signaling in astrocytes through primary cilia. Our in vivo study demonstrated that inhibition of either EV release or primary cilia prevents morphine tolerance in mice. CONCLUSIONS EV-mediated primary ciliogenesis contributes to the development of morphine tolerance.
Collapse
|
35
|
Kuthati Y, Rao VN, Busa P, Wong CS. Teneligliptin Exerts Antinociceptive Effects in Rat Model of Partial Sciatic Nerve Transection Induced Neuropathic Pain. Antioxidants (Basel) 2021; 10:antiox10091438. [PMID: 34573072 PMCID: PMC8465046 DOI: 10.3390/antiox10091438] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/26/2021] [Accepted: 09/07/2021] [Indexed: 01/13/2023] Open
Abstract
Neuropathic pain (NP), is a chronic pain resulting from nerve injury, with limited treatment options. Teneligliptin (TEN) is a dipeptidyl peptidase-4 inhibitor (DPP-4i) approved to treat type 2 diabetes. DPP-4is prevent the degradation of the incretin hormone glucagon-like peptide 1 (GLP-1) and prolong its circulation. Apart from glycemic control, GLP-1 is known to have antinociceptive and anti-inflammatory effects. Herein, we investigated the antinociceptive properties of TEN on acute pain, and partial sciatic nerve transection (PSNT)-induced NP in Wistar rats. Seven days post PSNT, allodynia and hyperalgesia were confirmed as NP, and intrathecal (i.t) catheters were implanted and connected to an osmotic pump for the vehicle (1 μL/h) or TEN (5 μg/1 μL/h) or TEN (5 μg) + GLP-1R antagonist Exendin-3 (9–39) amide (EXE) 0.1 μg/1 μL/h infusion. The tail-flick response, mechanical allodynia, and thermal hyperalgesia were measured for 7 more days. On day 14, the dorsal horn was harvested and used for Western blotting and immunofluorescence assays. The results showed that TEN had mild antinociceptive effects against acute pain but remarkable analgesic effects against NP. Furthermore, co-infusion of GLP-1R antagonist EXE with TEN partially reversed allodynia but not tail-flick latency. Immunofluorescence examination of the spinal cord revealed that TEN decreased the immunoreactivity of glial fibrillary acidic protein (GFAP). Taken together, our findings suggest that TEN is efficient in attenuation of PSNT-induced NP. Hence, the pleiotropic effects of TEN open a new avenue for NP management.
Collapse
Affiliation(s)
- Yaswanth Kuthati
- Department of Anesthesiology, Cathy General Hospital, Taipei 280, Taiwan; (Y.K.); (P.B.)
| | - Vaikar Navakanth Rao
- Department of Biomedical Sciences, Academia Sinica Institute, Taipei 11529, Taiwan;
| | - Prabhakar Busa
- Department of Anesthesiology, Cathy General Hospital, Taipei 280, Taiwan; (Y.K.); (P.B.)
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathy General Hospital, Taipei 280, Taiwan; (Y.K.); (P.B.)
- National Defense Medical Center, Institute of Medical Sciences, Taipei 280, Taiwan
- Correspondence: ; Tel.: +886-2-270-82-121; Fax: +886-2-879-24-835
| |
Collapse
|
36
|
Zhang L, Roy S. Opioid Modulation of the Gut-Brain Axis in Opioid-Associated Comorbidities. Cold Spring Harb Perspect Med 2021; 11:a040485. [PMID: 32816876 PMCID: PMC8415294 DOI: 10.1101/cshperspect.a040485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Growing evidence from animal and human studies show that opioids have a major impact on the composition and function of gut microbiota. This leads to disruption in gut permeability and altered microbial metabolites, driving both systemic and neuroinflammation, which in turn impacts central nervous system (CNS) homeostasis. Tolerance and dependence are the major comorbidities associated with prolonged opioid use. Inflammatory mediators and signaling pathways have been implicated in both opioid tolerance and dependence. We provide evidence that targeting the gut microbiome during opioid use through prebiotics, probiotics, antibiotics, and fecal microbial transplantation holds the greatest promise for novel treatments for opioid abuse. Basic research and clinical trials are required to examine what is more efficacious to yield new insights into the role of the gut-brain axis in opioid abuse.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology, University of Minnesota, Minnesota McGuire Translational Research Facility, Minneapolis, Minnesota 55455, USA
| | - Sabita Roy
- Department of Pharmacology, University of Minnesota, Minnesota McGuire Translational Research Facility, Minneapolis, Minnesota 55455, USA
- Department of Surgery, University of Miami, Miami, Florida 33153, USA
| |
Collapse
|
37
|
Wang T, Zhu X, Yi H, Gu J, Liu S, Izenwasser S, Lemmon VP, Roy S, Hao S. Viral vector-mediated gene therapy for opioid use disorders. Exp Neurol 2021; 341:113710. [PMID: 33781732 DOI: 10.1016/j.expneurol.2021.113710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
Chronic exposure to opioids typically results in adverse consequences. Opioid use disorder (OUD) is a disease of the CNS with behavioral, psychological, neurobiological, and medical manifestations. OUD induces a variety of changes of neurotransmitters/neuropeptides in the nervous system. Existing pharmacotherapy, such as opioid maintenance therapy (OMT) is the mainstay for the treatment of OUD, however, current opioid replacement therapy is far from effective for the majority of patients. Pharmacological therapy for OUD has been challenging for many reasons including debilitating side-effects. Therefore, developing an effective, non-pharmacological approach would be a critical advancement in improving and expanding treatment for OUD. Viral vector mediated gene therapy provides a potential new approach for treating opioid abused patients. Gene therapy can supply targeting gene products directly linked to the mechanisms of OUD to restore neurotransmitter and/or neuropeptides imbalance, and avoid the off-target effects of systemic administration of drugs. The most commonly used viral vectors in rodent studies of treatment of opioid-used disorder are based on recombinant adenovirus (AV), adeno-associated virus (AAV), lentiviral (LV) vectors, and herpes simplex virus (HSV) vectors. In this review, we will focus on the recent progress of viral vector mediated gene therapy in OUD, especially morphine tolerance and withdrawal.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Xun Zhu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hyun Yi
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Jun Gu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shue Liu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sari Izenwasser
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Vance P Lemmon
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shuanglin Hao
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America.
| |
Collapse
|
38
|
Lucerne KE, Osman A, Meckel KR, Kiraly DD. Contributions of neuroimmune and gut-brain signaling to vulnerability of developing substance use disorders. Neuropharmacology 2021; 192:108598. [PMID: 33965398 PMCID: PMC8220934 DOI: 10.1016/j.neuropharm.2021.108598] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023]
Abstract
Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aya Osman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
39
|
Wang Z, Jiang C, He Q, Matsuda M, Han Q, Wang K, Bang S, Ding H, Ko MC, Ji RR. Anti-PD-1 treatment impairs opioid antinociception in rodents and nonhuman primates. Sci Transl Med 2021; 12:12/531/eaaw6471. [PMID: 32075945 DOI: 10.1126/scitranslmed.aaw6471] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 07/15/2019] [Accepted: 12/19/2019] [Indexed: 12/31/2022]
Abstract
Emerging immunotherapies with monoclonal antibodies against programmed cell death protein-1 (PD-1) have shown success in treating cancers. However, PD-1 signaling in neurons is largely unknown. We recently reported that dorsal root ganglion (DRG) primary sensory neurons express PD-1 and activation of PD-1 inhibits neuronal excitability and pain. Opioids are mainstay treatments for cancer pain, and morphine produces antinociception via mu opioid receptor (MOR). Here, we report that morphine antinociception and MOR signaling require neuronal PD-1. Morphine-induced antinociception after systemic or intrathecal injection was compromised in Pd1 -/- mice. Morphine antinociception was also diminished in wild-type mice after intravenous or intrathecal administration of nivolumab, a clinically used anti-PD-1 monoclonal antibody. In mouse models of inflammatory, neuropathic, and cancer pain, spinal morphine antinociception was compromised in Pd1 -/- mice. MOR and PD-1 are coexpressed in sensory neurons and their axons in mouse and human DRG tissues. Morphine produced antinociception by (i) suppressing calcium currents in DRG neurons, (ii) suppressing excitatory synaptic transmission, and (iii) inducing outward currents in spinal cord neurons; all of these actions were impaired by PD-1 blockade in mice. Loss of PD-1 also enhanced opioid-induced hyperalgesia and tolerance and potentiates opioid-induced microgliosis and long-term potentiation in the spinal cord in mice. Last, intrathecal infusion of nivolumab inhibited intrathecal morphine-induced antinociception in nonhuman primates. Our findings demonstrate that PD-1 regulates opioid receptor signaling in nociceptive neurons, leading to altered opioid-induced antinociception in rodents and nonhuman primates.
Collapse
Affiliation(s)
- Zilong Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Qianru He
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Megumi Matsuda
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Qingjian Han
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kaiyuan Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sangsu Bang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Huiping Ding
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.,W. G. Hefner Veterans Affairs Medical Center, Salisbury, NC 28144, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA. .,Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
40
|
Su LY, Liu Q, Jiao L, Yao YG. Molecular Mechanism of Neuroprotective Effect of Melatonin on Morphine Addiction and Analgesic Tolerance: an Update. Mol Neurobiol 2021; 58:4628-4638. [PMID: 34148215 DOI: 10.1007/s12035-021-02448-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 06/07/2021] [Indexed: 12/22/2022]
Abstract
Drug addiction is a global health problem and continues to place an enormous financial burden on society. This addiction is characterized by drug dependence sensitization and craving. Morphine has been widely used for pain relief, but chronic administration of morphine causes analgesic tolerance, hyperalgesia, and addiction, all of which limit its clinical usage. Alterations of multiple molecular pathways have been reported to be involved in the development of drug addiction, including mitochondrial dysfunction, excessive oxidative stress and nitric oxide stress, and increased levels of apoptosis, autophagy, and neuroinflammation. Preclinical and clinical studies have shown that the co-administration of melatonin with morphine leads to a reversal of these affected pathways. In addition, murine models have shown that melatonin improves morphine-induced analgesic tolerance and addictive behaviors, such as behavioral sensitization, reward effect, and physical dependence. In this review, we attempt to summarize the recent findings about the beneficial effect and molecular mechanism of melatonin on mitochondrial dysfunction, uncontrolled autophagy, and neuroinflammation in morphine addiction and morphine analgesic tolerance. We propose that melatonin might be a useful supplement in the treatment opiate abuse.
Collapse
Affiliation(s)
- Ling-Yan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China. .,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China.
| | - Qianjin Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Lijin Jiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China. .,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
41
|
Li Y, Bao Y, Zheng H, Qin Y, Hua B. Can Src protein tyrosine kinase inhibitors be combined with opioid analgesics? Src and opioid-induced tolerance, hyperalgesia and addiction. Biomed Pharmacother 2021; 139:111653. [PMID: 34243625 DOI: 10.1016/j.biopha.2021.111653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022] Open
Abstract
The clinical application of opioids may be accompanied by a series of adverse consequences, such as opioid tolerance, opioid-induced hyperalgesia, opioid dependence or addiction. In view of this issue, clinicians are faced with the dilemma of treating various types of pain with or without opioids. In this review, we discuss that Src protein tyrosine kinase plays an important role in these adverse consequences, and Src inhibitors can solve these problems well. Therefore, Src inhibitors have the potential to be used in combination with opioids to achieve synergy. How to combine them together to maximize the analgesic effect while avoiding unnecessary trouble provides a topic for follow-up research.
Collapse
Affiliation(s)
- Yaoyuan Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yinggang Qin
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
42
|
Dos Reis Izolan L, da Silva DM, Oliveira HBL, de Oliveira Salomon JL, Peruzzi CP, Garcia SC, Dallegrave E, Zanotto C, Elisabetsky E, Gonçalves CA, Arbo MD, Konrath EL, Leal MB. Sintocalmy, a Passiflora incarnata Based Herbal, Attenuates Morphine Withdrawal in Mice. Neurochem Res 2021; 46:1092-1100. [PMID: 33544325 DOI: 10.1007/s11064-021-03237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 12/19/2020] [Accepted: 01/06/2021] [Indexed: 11/29/2022]
Abstract
Chronic opioid use changes brain chemistry in areas related to reward processes, memory, decision-making, and addiction. Both neurons and astrocytes are affected, ultimately leading to dependence. Passiflora incarnata L. (Passifloraceae) is the basis of frequently used herbals to manage anxiety and insomnia, with proven central nervous system depressant effects. Anti-addiction properties of P. incarnata have been reported. The aim of this study was to investigate the effect of a commercial extract of Passiflora incarnata (Sintocalmy®, Aché Laboratory) in the naloxone-induced jumping mice model of morphine withdrawal. In addition, glial fibrillary acidic protein (GFAP) and S100 calcium-binding protein B (S100B) levels were assessed in the frontal cortex and hippocampus, and DNA damage was verified on blood cells. In order to improve solubilization a Sintocalmy methanol extract (SME) was used. SME is mainly composed by flavonoids isovitexin and vitexin. The effects of SME 50, 100 and 200 mg/kg (i.p.) were evaluated in the naloxone-induced withdrawal syndrome in mice. SME 50 and SME 100 mg/kg decreased naloxone-induced jumping in morphine-dependent mice without reducing locomotor activity. No alterations were found in GFAP levels, however SME 50 mg/kg prevented the S100B increase in the frontal cortex and DNA damage. This study shows anti-addiction effects for a commercial standardized extract of P. incarnata and suggests the relevance of proper clinical assessment.
Collapse
Affiliation(s)
- Lucas Dos Reis Izolan
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Douglas Marques da Silva
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Helena Beatriz Larrosa Oliveira
- Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Janaína Lucas de Oliveira Salomon
- Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Caroline Portela Peruzzi
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Solange C Garcia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eliane Dallegrave
- Departamento de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Caroline Zanotto
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Elaine Elisabetsky
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Carlos Alberto Gonçalves
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Marcelo Dutra Arbo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo Luis Konrath
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mirna Bainy Leal
- Programa de Pós-Graduação em Ciências Biológicas - Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil. .,Laboratório de Farmacologia e Toxicologia de Produtos Naturais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
43
|
Fullerton EF, Rubaharan M, Karom MC, Hanberry RI, Murphy AZ. Advanced age attenuates the antihyperalgesic effect of morphine and decreases μ-opioid receptor expression and binding in the rat midbrain periaqueductal gray in male and female rats. Neurobiol Aging 2021; 98:78-87. [PMID: 33249376 PMCID: PMC8673746 DOI: 10.1016/j.neurobiolaging.2020.10.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/23/2020] [Accepted: 10/22/2020] [Indexed: 02/08/2023]
Abstract
The present study investigated the impact of advanced age on morphine modulation of persistent inflammatory pain in male and female rats. The impact of age, sex, and pain on μ-opioid receptor (MOR) expression and binding in the ventrolateral periaqueductal gray (vlPAG) was also examined using immunohistochemistry and receptor autoradiography. Intraplantar administration of complete Freund's adjuvant induced comparable levels of edema and hyperalgesia in adult (2-3 mos) and aged (16-18 mos) male and female rats. Morphine potency was highest in adult males, with a greater than two-fold increase in morphine EC50 observed in adult versus aged males (3.83 mg/kg vs. 10.16 mg/kg). Adult and aged female rats also exhibited significantly higher EC50 values (7.76 mg/kg and 8.74 mg/kg, respectively) than adult males. The upward shift in EC50 from adult to aged males was paralleled by a reduction in vlPAG MOR expression and binding. The observed age-related reductions in morphine potency and vlPAG MOR expression and binding have significant implications in pain management in the aged population.
Collapse
Affiliation(s)
- Evan F Fullerton
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | - Mary C Karom
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
44
|
Barbosa J, Faria J, Garcez F, Leal S, Afonso LP, Nascimento AV, Moreira R, Pereira FC, Queirós O, Carvalho F, Dinis-Oliveira RJ. Repeated Administration of Clinically Relevant Doses of the Prescription Opioids Tramadol and Tapentadol Causes Lung, Cardiac, and Brain Toxicity in Wistar Rats. Pharmaceuticals (Basel) 2021; 14:ph14020097. [PMID: 33513867 PMCID: PMC7912343 DOI: 10.3390/ph14020097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/19/2021] [Accepted: 01/23/2021] [Indexed: 12/18/2022] Open
Abstract
Tramadol and tapentadol, two structurally related synthetic opioid analgesics, are widely prescribed due to the enhanced therapeutic profiles resulting from the synergistic combination between μ-opioid receptor (MOR) activation and monoamine reuptake inhibition. However, the number of adverse reactions has been growing along with their increasing use and misuse. The potential toxicological mechanisms for these drugs are not completely understood, especially for tapentadol, owing to its shorter market history. Therefore, in the present study, we aimed to comparatively assess the putative lung, cardiac, and brain cortex toxicological damage elicited by the repeated exposure to therapeutic doses of both prescription opioids. To this purpose, male Wistar rats were intraperitoneally injected with single daily doses of 10, 25, and 50 mg/kg tramadol or tapentadol, corresponding to a standard analgesic dose, an intermediate dose, and the maximum recommended daily dose, respectively, for 14 consecutive days. Such treatment was found to lead mainly to lipid peroxidation and inflammation in lung and brain cortex tissues, as shown through augmented thiobarbituric acid reactive substances (TBARS), as well as to increased serum inflammation biomarkers, such as C reactive protein (CRP) and tumor necrosis factor-α (TNF-α). Cardiomyocyte integrity was also shown to be affected, since both opioids incremented serum lactate dehydrogenase (LDH) and α-hydroxybutyrate dehydrogenase (α-HBDH) activities, while tapentadol was associated with increased serum creatine kinase muscle brain (CK-MB) isoform activity. In turn, the analysis of metabolic parameters in brain cortex tissue revealed increased lactate concentration upon exposure to both drugs, as well as augmented LDH and creatine kinase (CK) activities following tapentadol treatment. In addition, pneumo- and cardiotoxicity biomarkers were quantified at the gene level, while neurotoxicity biomarkers were quantified both at the gene and protein levels; changes in their expression correlate with the oxidative stress, inflammatory, metabolic, and histopathological changes that were detected. Hematoxylin and eosin (H & E) staining revealed several histopathological alterations, including alveolar collapse and destruction in lung sections, inflammatory infiltrates, altered cardiomyocytes and loss of striation in heart sections, degenerated neurons, and accumulation of glial and microglial cells in brain cortex sections. In turn, Masson's trichrome staining confirmed fibrous tissue deposition in cardiac tissue. Taken as a whole, these results show that the repeated administration of both prescription opioids extends the dose range for which toxicological injury is observed to lower therapeutic doses. They also reinforce previous assumptions that tramadol and tapentadol are not devoid of toxicological risk even at clinical doses.
Collapse
Affiliation(s)
- Joana Barbosa
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence: (J.B.); (R.J.D.-O.); Tel.: +351-224-157-216 (J.B.); +351-224-157-216 (R.J.D.-O.)
| | - Juliana Faria
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Fernanda Garcez
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Sandra Leal
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- Department of Biomedicine, Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- CINTESIS—Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Luís Pedro Afonso
- Department of Pathology, Portuguese Institute of Oncology of Porto, 4200-072 Porto, Portugal;
| | - Ana Vanessa Nascimento
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Roxana Moreira
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Frederico C. Pereira
- Institute of Pharmacology and Experimental Therapeutics/iCBR, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal;
| | - Odília Queirós
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Félix Carvalho
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Ricardo Jorge Dinis-Oliveira
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence: (J.B.); (R.J.D.-O.); Tel.: +351-224-157-216 (J.B.); +351-224-157-216 (R.J.D.-O.)
| |
Collapse
|
45
|
Alifarsangi A, Esmaeili-Mahani S, Sheibani V, Abbasnejad M. The citrus flavanone naringenin prevents the development of morphine analgesic tolerance and conditioned place preference in male rats. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2021; 47:43-51. [PMID: 33006902 DOI: 10.1080/00952990.2020.1813296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Opioids are effective analgesics in the management of chronic pain. However, their clinical use is hindered by adverse side effects such as addiction and analgesic tolerance. Naringenin is a common polyphenolic constituent of the citrus fruits and is one of the most commonly consumed flavonoids within our regular diet. However, its influences on opioid tolerance and addiction have not yet been clarified. OBJECTIVES To examine the effect of different doses of naringenin on analgesic tolerance, conditioned place preference and neuroinflammation in morphine-exposed rats. METHODS Analgesic tolerance was induced by the injection of 10 mg/kg morphine twice daily for 8 days in 70 male Wistar rats. To evaluate the effect of naringenin on the development of morphine tolerance, different doses (10, 25 and 50 mg/kg i.p.) were injected 15 min before morphine. The tail-flick test was used to assess nociceptive threshold. Conditioned place preference test was used to evaluate morphine-seeking behaviors. The lumbar spinal cord was assayed to determine glial fibrillary acidic protein (GFAP) and cyclooxygenase-2 (COX-2) levels by Western blotting. RESULTS The data showed that naringenin could significantly prevent morphine tolerance (p < .001) and conditioned place preference. In addition, chronic morphine-induced GFAP and COX-2 overexpression was significantly reversed by 50 mg/kg naringenin (p < .05 and p < .01, respectively). CONCLUSION Our results suggest that naringenin may have a potential anti-tolerant/anti-addiction property against chronic morphine misuse and that this preventive effect is associated with its anti-neuroinflammatory effects.
Collapse
Affiliation(s)
- Atena Alifarsangi
- Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Saeed Esmaeili-Mahani
- Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Vahid Sheibani
- Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Abbasnejad
- Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
46
|
Koob GF. Drug Addiction: Hyperkatifeia/Negative Reinforcement as a Framework for Medications Development. Pharmacol Rev 2021; 73:163-201. [PMID: 33318153 PMCID: PMC7770492 DOI: 10.1124/pharmrev.120.000083] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Compulsive drug seeking that is associated with addiction is hypothesized to follow a heuristic framework that involves three stages (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) and three domains of dysfunction (incentive salience/pathologic habits, negative emotional states, and executive function, respectively) via changes in the basal ganglia, extended amygdala/habenula, and frontal cortex, respectively. This review focuses on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the addiction cycle. Hyperkatifeia provides an additional source of motivation for compulsive drug seeking via negative reinforcement. Negative reinforcement reflects an increase in the probability of a response to remove an aversive stimulus or drug seeking to remove hyperkatifeia that is augmented by genetic/epigenetic vulnerability, environmental trauma, and psychiatric comorbidity. Neurobiological targets for hyperkatifeia in addiction involve neurocircuitry of the extended amygdala and its connections via within-system neuroadaptations in dopamine, enkephalin/endorphin opioid peptide, and γ-aminobutyric acid/glutamate systems and between-system neuroadaptations in prostress corticotropin-releasing factor, norepinephrine, glucocorticoid, dynorphin, hypocretin, and neuroimmune systems and antistress neuropeptide Y, nociceptin, endocannabinoid, and oxytocin systems. Such neurochemical/neurocircuitry dysregulations are hypothesized to mediate a negative hedonic set point that gradually gains allostatic load and shifts from a homeostatic hedonic state to an allostatic hedonic state. Based on preclinical studies and translational studies to date, medications and behavioral therapies that reset brain stress, antistress, and emotional pain systems and return them to homeostasis would be promising new targets for medication development. SIGNIFICANCE STATEMENT: The focus of this review is on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the drug addiction cycle and a driving force for negative reinforcement in addiction. Medications and behavioral therapies that reverse hyperkatifeia by resetting brain stress, antistress, and emotional pain systems and returning them to homeostasis would be promising new targets for medication development.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
47
|
Hahm SC, Lee JS, Yoon YW, Kim J. Analgesic Tolerance Development during Repetitive Electric Stimulations Is Associated with Changes in the Expression of Activated Microglia in Rats with Osteoarthritis. Biomedicines 2020; 8:biomedicines8120575. [PMID: 33297333 PMCID: PMC7762208 DOI: 10.3390/biomedicines8120575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/01/2020] [Accepted: 12/05/2020] [Indexed: 11/30/2022] Open
Abstract
Electric stimulation is used for managing osteoarthritic (OA) pain; however, little is known about the development of analgesic tolerance during repeated stimulations and the relation of spinal microglia with OA pain. We investigated the changes in the analgesic effects of repeated electric stimulations and the relation between the development of analgesic tolerance and spinal microglial expression in rats with OA. To induce OA, monosodium iodoacetate was injected into the synovial space of the right knee joint of the rats (n = 185). Repeated high frequency, low frequency, or sham transcutaneous electric nerve stimulation (TENS) was performed to the ipsilateral knee joint for 20 min in rats with OA (n = 45). Minocycline or minocycline plus TENS (HF, LF, or sham) was treated in OA rats with repeated TENS-induced tolerance (n = 135). Immunohistochemistry of the microglia in the L3–L5 spinal segments was performed. Knee joint pain during passive movement of the knee joint were quantified using the knee-bend score and the proportion of activated microglia was calculated as primary variables. Paw withdrawal threshold (hypersensitivity to mechanical stimuli) was assessed and the resting and activated microglia were counted as secondary variables. Repeated applications decreased the analgesic effect of TENS on OA pain and failed to reduce the expression of activated microglia in the spinal cord. However, spinal microglial inhibition by minocycline restored the analgesic effect of TENS on OA pain in TENS-tolerant OA rats. TENS combined with minocycline treatment improved knee joint pain and mechanical hypersensitivity in TENS-tolerant OA rats, and inhibited the expression of activated microglia in the spinal cord. These results suggest a possible relationship between repetitive electric stimulation-induced analgesic tolerance for OA pain control and changes in microglia activation.
Collapse
Affiliation(s)
- Suk-Chan Hahm
- Graduate School of Integrative Medicine, CHA University, Seongnam 13488, Korea;
| | - Jin Seung Lee
- Department of Physical Therapy, Korea University College of Health Science, Seoul 02841, Korea;
- Rehabilitation Science Program, Department of Health Sciences, Graduate School, Korea University, Seoul 02841, Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul 02841, Korea
| | - Young Wook Yoon
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Korea;
| | - Junesun Kim
- Department of Physical Therapy, Korea University College of Health Science, Seoul 02841, Korea;
- Rehabilitation Science Program, Department of Health Sciences, Graduate School, Korea University, Seoul 02841, Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul 02841, Korea
- Correspondence: ; Tel.: +82-2-3290-5689; Fax: +82-2-921-7260
| |
Collapse
|
48
|
Linher-Melville K, Shah A, Singh G. Sex differences in neuro(auto)immunity and chronic sciatic nerve pain. Biol Sex Differ 2020; 11:62. [PMID: 33183347 PMCID: PMC7661171 DOI: 10.1186/s13293-020-00339-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/20/2020] [Indexed: 01/13/2023] Open
Abstract
Chronic pain occurs with greater frequency in women, with a parallel sexually dimorphic trend reported in sufferers of many autoimmune diseases. There is a need to continue examining neuro-immune-endocrine crosstalk in the context of sexual dimorphisms in chronic pain. Several phenomena in particular need to be further explored. In patients, autoantibodies to neural antigens have been associated with sensory pathway hyper-excitability, and the role of self-antigens released by damaged nerves remains to be defined. In addition, specific immune cells release pro-nociceptive cytokines that directly influence neural firing, while T lymphocytes activated by specific antigens secrete factors that either support nerve repair or exacerbate the damage. Modulating specific immune cell populations could therefore be a means to promote nerve recovery, with sex-specific outcomes. Understanding biological sex differences that maintain, or fail to maintain, neuroimmune homeostasis may inform the selection of sex-specific treatment regimens, improving chronic pain management by rebalancing neuroimmune feedback. Given the significance of interactions between nerves and immune cells in the generation and maintenance of neuropathic pain, this review focuses on sex differences and possible links with persistent autoimmune activity using sciatica as an example.
Collapse
Affiliation(s)
- Katja Linher-Melville
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| | - Anita Shah
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
49
|
Lucerne KE, Kiraly DD. The role of gut-immune-brain signaling in substance use disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:311-370. [PMID: 33648673 DOI: 10.1016/bs.irn.2020.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Substance use disorders (SUDs) are debilitating neuropsychiatric conditions that exact enormous costs in terms of loss of life and individual suffering. While much progress has been made defining the neurocircuitry and intracellular signaling cascades that contribute to SUDs, these studies have yielded limited effective treatment options. This has prompted greater exploration of non-traditional targets in addiction. Emerging data suggest inputs from peripheral systems, such as the immune system and the gut microbiome, impact multiple neuropsychiatric diseases, including SUDs. Until recently the gut microbiome, peripheral immune system, and the CNS have been studied independently; however, current work shows the gut microbiome and immune system critically interact to modulate brain function. Additionally, the gut microbiome and immune system intimately regulate one another via extensive bidirectional communication. Accumulating evidence suggests an important role for gut-immune-brain communication in the pathogenesis of substance use disorders. Thus, a better understanding of gut-immune-brain signaling could yield important insight to addiction pathology and potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
50
|
Doyle TM, Hutchinson MR, Braden K, Janes K, Staikopoulos V, Chen Z, Neumann WL, Spiegel S, Salvemini D. Sphingosine-1-phosphate receptor subtype 1 activation in the central nervous system contributes to morphine withdrawal in rodents. J Neuroinflammation 2020; 17:314. [PMID: 33092620 PMCID: PMC7584082 DOI: 10.1186/s12974-020-01975-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/30/2020] [Indexed: 01/08/2023] Open
Abstract
Opioid therapies for chronic pain are undermined by many adverse side effects that reduce their efficacy and lead to dependence, abuse, reduced quality of life, and even death. We have recently reported that sphingosine-1-phosphate (S1P) 1 receptor (S1PR1) antagonists block the development of morphine-induced hyperalgesia and analgesic tolerance. However, the impact of S1PR1 antagonists on other undesirable side effects of opioids, such as opioid-induced dependence, remains unknown. Here, we demonstrate that naloxone-precipitated morphine withdrawal in mice altered de novo sphingolipid metabolism in the dorsal horn of the spinal cord and increased S1P that accompanied the manifestation of several withdrawal behaviors. Blocking de novo sphingolipid metabolism with intrathecal administration of myriocin, an inhibitor of serine palmitoyltransferase, blocked naloxone-precipitated withdrawal. Noteworthy, we found that competitive (NIBR-15) and functional (FTY720) S1PR1 antagonists attenuated withdrawal behaviors in mice. Mechanistically, at the level of the spinal cord, naloxone-precipitated withdrawal was associated with increased glial activity and formation of the potent inflammatory/neuroexcitatory cytokine interleukin-1β (IL-1β); these events were attenuated by S1PR1 antagonists. These results provide the first molecular insight for the role of the S1P/S1PR1 axis during opioid withdrawal. Our data identify S1PR1 antagonists as potential therapeutics to mitigate opioid-induced dependence and support repurposing the S1PR1 functional antagonist FTY720, which is FDA-approved for multiple sclerosis, as an opioid adjunct.
Collapse
Affiliation(s)
- Timothy M Doyle
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA.,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA
| | - Mark R Hutchinson
- Discipline of Physiology, University of Adelaide, Adelaide, South Australia, 5005, Australia.,Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, South Australia, 5005, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Kathryn Braden
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA.,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA
| | - Kali Janes
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA
| | - Vicky Staikopoulos
- Discipline of Physiology, University of Adelaide, Adelaide, South Australia, 5005, Australia.,Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, South Australia, 5005, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Zhoumou Chen
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA.,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA
| | - William L Neumann
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, 200 University Park, Edwardsville, IL, 62026, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine, 1101 E Marshall St, Richmond, VA, 23298, USA
| | - Daniela Salvemini
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA. .,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA.
| |
Collapse
|