1
|
Sommer G, Rodríguez López C, Hirschkorn A, Calimano G, Marques-Lopes J, Milner TA, Glass MJ. Estrogen Receptor Beta Agonist Influences Presynaptic NMDA Receptor Distribution in the Paraventricular Hypothalamic Nucleus Following Hypertension in a Mouse Model of Perimenopause. BIOLOGY 2024; 13:819. [PMID: 39452127 PMCID: PMC11505520 DOI: 10.3390/biology13100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/01/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Women become susceptible to hypertension as they transition to menopause (i.e., perimenopause); however, the underlying mechanisms are unclear. Animal studies using an accelerated ovarian failure (AOF) model of peri-menopause (peri-AOF) demonstrate that peri-AOF hypertension is associated with increased postsynaptic NMDA receptor plasticity in the paraventricular hypothalamic nucleus (PVN), a brain area critical for blood pressure regulation. However, recent evidence indicates that presynaptic NMDA receptors also play a role in neural plasticity. Here, using immuno-electron microscopy, we examine the influence of peri-AOF hypertension on the subcellular distribution of the essential NMDA GluN1 receptor subunit in PVN axon terminals in peri-AOF and in male mice. Hypertension was produced by 14-day slow-pressor angiotensin II (AngII) infusion. The involvement of estrogen signaling was investigated by co-administering an estrogen receptor beta (ERß) agonist. Although AngII induced hypertension in both peri-AOF and male mice, peri-AOF females showed higher cytoplasmic GluN1 levels. In peri-AOF females, activation of ERß blocked hypertension and increased plasmalemmal GluN1 in axon terminals. In contrast, stimulation of ERß did not inhibit hypertension or influence presynaptic GluN1 localization in males. These results indicate that sex-dependent recruitment of presynaptic NMDA receptors in the PVN is influenced by ERß signaling in mice during early ovarian failure.
Collapse
Affiliation(s)
- Garrett Sommer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Claudia Rodríguez López
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Adi Hirschkorn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Gianna Calimano
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Jose Marques-Lopes
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
- Center for Translational Health and Medical Biotechnology Research (TBIO)/Health Research Network (RISE-HEALTH), ESS, Polytechnic of Porto, R. Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| | - Michael J. Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA (C.R.L.); (J.M.-L.)
| |
Collapse
|
2
|
Abstract
The harmful side effects of opioid drugs such as respiratory depression, tolerance, dependence, and abuse potential have limited the therapeutic utility of opioids for their entire clinical history. However, no previous attempt to develop effective pain drugs that substantially ameliorate these effects has succeeded, and the current opioid epidemic affirms that they are a greater hindrance to the field of pain management than ever. Recent attempts at new opioid development have sought to reduce these side effects by minimizing engagement of the regulatory protein arrestin-3 at the mu-opioid receptor, but there is significant controversy around this approach. Here, we discuss the ongoing effort to develop safer opioids and its relevant historical context. We propose a new model that reconciles results previously assumed to be in direct conflict to explain how different signaling profiles at the mu-opioid receptor contribute to opioid tolerance and dependence. Our goal is for this framework to inform the search for a new generation of lower liability opioid analgesics.
Collapse
Affiliation(s)
| | - Jennifer L Whistler
- Center for Neuroscience, University of California, Davis, California, USA;
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, California, USA
| |
Collapse
|
3
|
Ochandarena NE, Niehaus JK, Tassou A, Scherrer G. Cell-type specific molecular architecture for mu opioid receptor function in pain and addiction circuits. Neuropharmacology 2023; 238:109597. [PMID: 37271281 PMCID: PMC10494323 DOI: 10.1016/j.neuropharm.2023.109597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/13/2023] [Indexed: 06/06/2023]
Abstract
Opioids are potent analgesics broadly used for pain management; however, they can produce dangerous side effects including addiction and respiratory depression. These harmful effects have led to an epidemic of opioid abuse and overdose deaths, creating an urgent need for the development of both safer pain medications and treatments for opioid use disorders. Both the analgesic and addictive properties of opioids are mediated by the mu opioid receptor (MOR), making resolution of the cell types and neural circuits responsible for each of the effects of opioids a critical research goal. Single-cell RNA sequencing (scRNA-seq) technology is enabling the identification of MOR-expressing cell types throughout the nervous system, creating new opportunities for mapping distinct opioid effects onto newly discovered cell types. Here, we describe molecularly defined MOR-expressing neuronal cell types throughout the peripheral and central nervous systems and their potential contributions to opioid analgesia and addiction.
Collapse
Affiliation(s)
- Nicole E Ochandarena
- Neuroscience Curriculum, Biological and Biomedical Sciences Program, The University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Jesse K Niehaus
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
4
|
Sun Q, Li Z, Wang Z, Wang Q, Qin F, Pan H, Lin W, Mu X, Wang Y, Jiang Y, Ji J, Lu Z. Immunosuppression by opioids: Mechanisms of action on innate and adaptive immunity. Biochem Pharmacol 2023; 209:115417. [PMID: 36682388 DOI: 10.1016/j.bcp.2023.115417] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/22/2023]
Abstract
Opioids are excellent analgesics for the clinical treatment of various types of acute and chronic pain, particularly cancer-related pain. Nevertheless, it is well known that opioids have some nasty side effects, including immunosuppression, which is commonly overlooked. As a result, the incidence of opportunistic bacterial and viral infections increases in patients with long-term opioid use. Nowadays, there are no effective medications to alleviate opioid-induced immunosuppression. Understanding the underlying molecular mechanism of opioids in immunosuppression can enable researchers to devise effective therapeutic interventions. This review comprehensively summarized the exogenous opioids-induced immunosuppressive effects and their underlying mechanisms, the regulatory roles of endogenous opioids on the immune system, the potential link between opioid immunosuppressive effect and the function of the central nervous system (CNS), and the future perspectives in this field.
Collapse
Affiliation(s)
- Qinmei Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhonghao Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zijing Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qisheng Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Fenfen Qin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Haotian Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weixin Lin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xinru Mu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuxuan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yongwei Jiang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhigang Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
5
|
Dolgetta A, Johnson M, Fruitman K, Siegel L, Zhou Y, McEwen BS, Kreek MJ, Milner TA. Sex and chronic stress alter the distribution of glutamate receptors within rat hippocampal CA3 pyramidal cells following oxycodone conditioned place preference. Neurobiol Stress 2022; 17:100431. [PMID: 35535260 PMCID: PMC9076964 DOI: 10.1016/j.ynstr.2022.100431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/02/2022] [Accepted: 01/19/2022] [Indexed: 12/01/2022] Open
Abstract
Glutamate receptors have a key role in the neurobiology of opioid addiction. Using electron microscopic immunocytochemical methods, this project elucidates how sex and chronic immobilization stress (CIS) impact the redistribution of GluN1 and GluA1 within rat hippocampal CA3 pyramidal cells following oxycodone (Oxy) conditioned place preference (CPP). Four groups of female and male Sprague-Dawley rats subjected to CPP were used: Saline- (Sal) and Oxy-injected (3 mg/kg, I.P.) naïve rats; and Sal- and Oxy-injected CIS rats. GluN1: In both naive and CIS rats, Sal-females compared to Sal-males had elevated cytoplasmic and total dendritic GluN1. Following Oxy CPP, near plasmalemmal, cytoplasmic, and total GluN1 decreased in CA3 dendrites of unstressed females suggesting reduced pools of GluN1 available for ligand binding. Following CIS, Oxy-males (which did not acquire CPP) had increased GluN1 in all compartments of dendrites and spines of CA3 neurons. GluA1: There were no differences in the distribution GluA1 in any cellular compartments of CA3 dendrites in naïve females and males following either Sal or Oxy CPP. CIS alone increased the percent of GluA1 in CA3 dendritic spines in males compared to females. CIS Oxy-males compared to CIS Sal-males had an increase in cytoplasmic and total dendritic GluA1. Thus, in CIS Oxy-males increased pools of GluN1 and GluA1 are available for ligand binding in CA3 neurons. Together with our prior experiments, these changes in GluN1 and GluA1 following CIS in males may contribute to an increased sensitivity of CA3 neurons to glutamate excitation and a reduced capacity to acquire Oxy CPP.
Collapse
Key Words
- ABC, avidin-biotin complex
- AMPA receptors
- BSA, bovine serum albumin
- CIS, chronic immobilization stress
- CPP, conditioned place preference
- DAB, diaminobenzidine
- DG, dentate gyrus
- DOR, delta opioid receptor
- Drug associative-learning
- Electron microscopy
- GABA, Gamma-amino butyric acid
- GluA1, AMPA glutamate receptor subunit 1
- GluN1, NMDA, glutamate receptor subunit 1
- LTP, long-term potentiation
- MOR, mu opioid receptor
- NMDA receptors
- NMDA, N-methyl-D-aspartate
- NPY, neuropeptide Y
- Oxy, oxycodone
- PARV, parvalbumin
- PB, phosphate buffer
- PFA, paraformaldehyde
- PM, plasma membrane
- Pyramidal cells
- ROI, region of interest
- SLM, stratum lacunosum-moleculare
- SLu, stratum lucidum
- SO, stratum oriens
- SOM, somatostatin
- SR, stratum radiatum
- Sal, saline
- TS, tris-buffered saline
- ir, immunoreactivity
Collapse
Affiliation(s)
- Alexandra Dolgetta
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Megan Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Kate Fruitman
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Luke Siegel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
6
|
Sex differences in the rodent hippocampal opioid system following stress and oxycodone associated learning processes. Pharmacol Biochem Behav 2022; 212:173294. [PMID: 34752798 PMCID: PMC8748406 DOI: 10.1016/j.pbb.2021.173294] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 01/03/2023]
Abstract
Over the past two decades, opioid abuse has risen especially among women. In both sexes hippocampal neural circuits involved in associative memory formation and encoding of motivational incentives are critically important in the transition from initial drug use to drug abuse/dependence. Opioid circuits, particularly the mossy fiber pathway, are crucial for associative memory processes important for addiction. Our anatomical studies, especially those utilizing electron microscopic immunocytochemistry, have provided unique insight into sex differences in the distribution of opioid peptides and receptors in specific hippocampal circuits and how these distributions are altered following stress and oxycodone-associative learning processes. Here we review the hippocampal opioid system in rodents with respect to ovarian hormones effects and baseline sex differences then sex differences following acute and chronic stress. Next, we review sex differences in the hippocampal opioid system in unstressed and chronically stressed rats following oxycodone conditioned place preference. We show that opioid peptides and receptors are distributed within hippocampal circuits in females with elevated estrogen states in a manner that would enhance sensitivity to endogenous and exogenous opioids. Moreover, chronic stress primes the opioid system in females in a manner that would promote opioid-associative learning processes. In contrast, chronic stress has limited effects on the opioid system in males and reduces its capacity to support opioid-mediated learning processes. Interestingly, acute stress appears to prime males for opioid associative learning. On a broader scale the findings highlighted in this review have important implications in understanding sex differences in opioid drug use and abuse.
Collapse
|
7
|
Windisch KA, Mazid S, Johnson MA, Ashirova E, Zhou Y, Gergoire L, Warwick S, McEwen BS, Kreek MJ, Milner TA. Acute Delta 9-tetrahydrocannabinol administration differentially alters the hippocampal opioid system in adult female and male rats. Synapse 2021; 75:e22218. [PMID: 34255372 DOI: 10.1002/syn.22218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/22/2022]
Abstract
Our prior studies demonstrated that the rat hippocampal opioid system can undergo sex-specific adaptations to external stimuli that can influence opioid-associated learning processes. This opioid system extensively overlaps with the cannabinoid system. Moreover, acute administration of Δ9 Tetrahydrocannabinoid (THC), the primary psychoactive constituent of cannabis, can alter cognitive behaviors that involve the hippocampus. Here, we use light and electron microscopic immunocytochemical methods to examine the effects of acute THC (5 mg/kg, i.p., 1 h) on mossy fiber Leu-Enkephalin (LEnk) levels and the distribution and phosphorylation levels of delta and mu opioid receptors (DORs and MORs, respectively) in CA3 pyramidal cells and parvalbumin dentate hilar interneurons of adult female and male Sprague-Dawley rats. In females with elevated estrogen states (proestrus/estrus stage), acute THC altered the opioid system so that it resembled that seen in vehicle-injected females with low estrogen states (diestrus) and males: (1) mossy fiber LEnk levels in CA2/3a decreased; (2) phosphorylated-DOR levels in CA2/3a pyramidal cells increased; and (3) phosphorylated-MOR levels increased in most CA3b laminae. In males, acute THC resulted in the internalization of MORs in parvalbumin-containing interneuron dendrites which would decrease disinhibition of granule cells. In both sexes, acute THC redistributed DORs to the near plasma membrane of CA3 pyramidal cell dendrites, however, the dendritic region varied with sex. Additionally, acute THC also resulted in a sex-specific redistribution of DORs within CA3 pyramidal cell dendrites which could differentially promote synaptic plasticity and/or opioid-associated learning processes in both females and males.
Collapse
Affiliation(s)
- Kyle A Windisch
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Sanoara Mazid
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York
| | - Megan A Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York
| | - Elina Ashirova
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Lennox Gergoire
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York
| | - Sydney Warwick
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Teresa A Milner
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York
| |
Collapse
|
8
|
Rubin BR, Johnson MA, Berman JM, Goldstein E, Pertsovskaya V, Zhou Y, Contoreggi NH, Dyer AG, Gray JD, Waters EM, McEwen BS, Kreek MJ, Milner TA. Sex and chronic stress alter delta opioid receptor distribution within rat hippocampal CA1 pyramidal cells following behavioral challenges. Neurobiol Stress 2020; 13:100236. [PMID: 33344692 PMCID: PMC7739044 DOI: 10.1016/j.ynstr.2020.100236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
Following oxycodone (Oxy) conditioned place preference (CPP), delta opioid receptors (DORs) differentially redistribute in hippocampal CA3 pyramidal cells in female and male rats in a manner that would promote plasticity and opioid-associative learning processes. However, following chronic immobilization stress (CIS), males do not acquire Oxy-CPP and the trafficking of DORs in CA3 pyramidal neurons is attenuated. Here, we examined the subcellular distribution of DORs in CA1 pyramidal cells using electron microscopy in these same cohorts. CPP Saline (Sal)-females compared to Sal-males have more cytoplasmic and total DORs in dendrites and more DOR-labeled spines. Following Oxy-CPP, DORs redistribute from near-plasmalemma pools in dendrites to spines in males. CIS Control females compared to control males have more near-plasmalemmal dendritic DORs. Following CIS, dendritic DORs are elevated in the cytoplasm in females and near-plasmalemma in males. CIS plus CPP CIS Sal-females compared to CIS Sal-males have more DORs on the plasmalemma of dendrites and in spines. After Oxy, the distribution of DORs does not change in either females or males. Conclusion Following Oxy-CPP, DORs within CA1 pyramidal cells remain positioned in naïve female rats to enhance sensitivity to DOR agonists and traffic to dendritic spines in naïve males where they can promote plasticity processes. Following CIS plus behavioral enrichment, DORs are redistributed within CA1 pyramidal cells in females in a manner that could enhance sensitivity to DOR agonists. Conversely, CIS plus behavioral enrichment does not alter DORs in CA1 pyramidal cells in males, which may contribute to their diminished capacity to acquire Oxy-CPP.
Collapse
Affiliation(s)
- Batsheva R. Rubin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Megan A. Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Jared M. Berman
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Ellen Goldstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Vera Pertsovskaya
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Natalina H. Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Andreina G. Dyer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Jason D. Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Elizabeth M. Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
- Corresponding author. Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, RM 307 New York, NY 10065, United States.
| |
Collapse
|
9
|
Starnowska-Sokół J, Piotrowska A, Bogacka J, Makuch W, Mika J, Witkowska E, Godlewska M, Osiejuk J, Gątarz S, Misicka A, Przewłocka B. Novel hybrid compounds, opioid agonist+melanocortin 4 receptor antagonist, as efficient analgesics in mouse chronic constriction injury model of neuropathic pain. Neuropharmacology 2020; 178:108232. [PMID: 32750445 DOI: 10.1016/j.neuropharm.2020.108232] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/24/2020] [Accepted: 07/06/2020] [Indexed: 12/30/2022]
Abstract
When the nerve tissue is injured, endogenous agonist of melanocortin type 4 (MC4) receptor, α-MSH, exerts tonic pronociceptive action in the central nervous system, contributing to sustaining the neuropathic pain state and counteracting the analgesic effects of exogenous opioids. With the intent of enhancing opioid analgesia in neuropathy by blocking the MC4 activation, so-called parent compounds (opioid agonist, MC4 antagonist) were joined together using various linkers to create novel bifunctional hybrid compounds. Analgesic action of four hybrids was tested after intrathecal (i.t.) administration in mouse models of acute and neuropathic pain (chronic constriction injury model, CCI). Under nerve injury conditions, one of the hybrids, UW3, induced analgesia in 1500 times lower i.t. dose than the opioid parent (ED50: 0.0002 nmol for the hybrid, 0.3 nmol for the opioid parent) and in an over 16000 times lower dose than the MC4 parent (ED50: 3.33 nmol) as measured by the von Frey test. Two selected hybrids were tested for analgesic properties in CCI mice after intravenous (i.v.) and intraperitoneal (i.p.) administration. Opioid receptor antagonists and MC4 receptor agonists diminished the analgesic action of these two hybrids studied, though the extent of this effect differed between the hybrids; this suggests that linker is of key importance here. Further results indicate a significant advantage of hybrid compounds over the physical mixture of individual pharmacophores in their analgesic effect. All this evidence justifies the idea of synthesizing a bifunctional opioid agonist-linker-MC4 antagonist compound, as such structure may bring important benefits in neuropathic pain treatment.
Collapse
Affiliation(s)
- Joanna Starnowska-Sokół
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Anna Piotrowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Joanna Bogacka
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Wioletta Makuch
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Joanna Mika
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland
| | - Ewa Witkowska
- University of Warsaw, Faculty of Chemistry, Biological and Chemistry Research Centre, Warsaw, Poland
| | - Magda Godlewska
- University of Warsaw, Faculty of Chemistry, Biological and Chemistry Research Centre, Warsaw, Poland
| | - Jowita Osiejuk
- University of Warsaw, Faculty of Chemistry, Biological and Chemistry Research Centre, Warsaw, Poland
| | - Sandra Gątarz
- University of Warsaw, Faculty of Chemistry, Biological and Chemistry Research Centre, Warsaw, Poland
| | - Aleksandra Misicka
- University of Warsaw, Faculty of Chemistry, Biological and Chemistry Research Centre, Warsaw, Poland
| | - Barbara Przewłocka
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, Krakow, Poland.
| |
Collapse
|
10
|
Puig S, Barker KE, Szott SR, Kann PT, Morris JS, Gutstein HB. Spinal Opioid Tolerance Depends upon Platelet-Derived Growth Factor Receptor- β Signaling, Not μ-Opioid Receptor Internalization. Mol Pharmacol 2020; 98:487-496. [PMID: 32723769 PMCID: PMC7562976 DOI: 10.1124/mol.120.119552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/22/2020] [Indexed: 12/22/2022] Open
Abstract
Opioids are some of the most potent analgesics available. However, their effectiveness is limited by the development of analgesic tolerance. Traditionally, tolerance was thought to occur by termination of μ-opioid receptor (MOR) signaling via desensitization and internalization. Contradictory findings led to a more recent proposal that sustained MOR signaling caused analgesic tolerance. However, this view has also been called into question. We recently discovered that the platelet-derived growth factor receptor(PDGFR)-β signaling system is both necessary and sufficient to cause opioid tolerance. We therefore propose a completely new hypothesis: that opioid tolerance is mediated by selective cellular signals and is independent of MOR internalization. To test this hypothesis, we developed an automated software-based method to perform unbiased analyses of opioid-induced MOR internalization in the rat substantia gelatinosa. We induced tolerance with either morphine, which did not cause MOR internalization, or fentanyl, which did. We also blocked tolerance by administering morphine or fentanyl with the PDGFR-β inhibitor imatinib. We found that imatinib blocked tolerance without altering receptor internalization induced by either morphine or fentanyl. We also showed that imatinib blocked tolerance to other clinically used opioids. Our findings indicate that opioid tolerance is not dependent upon MOR internalization and support the novel hypothesis that opioid tolerance is mediated by intracellular signaling that can be selectively targeted. This suggests the exciting possibility that undesirable opioid side effects can be selectively eliminated, dramatically improving the safety and efficacy of opioids. SIGNIFICANCE STATEMENT: Classically, it was thought that analgesic tolerance to opioids was caused by desensitization and internalization of μ-opioid receptors (MORs). More recently, it was proposed that sustained, rather than reduced, MOR signaling caused tolerance. Here, we present conclusive evidence that opioid tolerance occurs independently of MOR internalization and that it is selectively mediated by platelet-derived growth factor receptor signaling. This novel hypothesis suggests that dangerous opioid side effects can be selectively targeted and blocked, improving the safety and efficacy of opioids.
Collapse
Affiliation(s)
- S Puig
- Anesthesiology Institute, Allegheny Health Network, Pittsburgh, Pennsylvania (H.B.G.); University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (S.P., S.R.S., P.T.K.); MD Anderson Cancer Center, Houston, Texas (K.E.B.); and Biostatistics Division, Perelman School of Medicine, Philadelphia, Pennsylvania (J.S.M.)
| | - K E Barker
- Anesthesiology Institute, Allegheny Health Network, Pittsburgh, Pennsylvania (H.B.G.); University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (S.P., S.R.S., P.T.K.); MD Anderson Cancer Center, Houston, Texas (K.E.B.); and Biostatistics Division, Perelman School of Medicine, Philadelphia, Pennsylvania (J.S.M.)
| | - S R Szott
- Anesthesiology Institute, Allegheny Health Network, Pittsburgh, Pennsylvania (H.B.G.); University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (S.P., S.R.S., P.T.K.); MD Anderson Cancer Center, Houston, Texas (K.E.B.); and Biostatistics Division, Perelman School of Medicine, Philadelphia, Pennsylvania (J.S.M.)
| | - P T Kann
- Anesthesiology Institute, Allegheny Health Network, Pittsburgh, Pennsylvania (H.B.G.); University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (S.P., S.R.S., P.T.K.); MD Anderson Cancer Center, Houston, Texas (K.E.B.); and Biostatistics Division, Perelman School of Medicine, Philadelphia, Pennsylvania (J.S.M.)
| | - J S Morris
- Anesthesiology Institute, Allegheny Health Network, Pittsburgh, Pennsylvania (H.B.G.); University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (S.P., S.R.S., P.T.K.); MD Anderson Cancer Center, Houston, Texas (K.E.B.); and Biostatistics Division, Perelman School of Medicine, Philadelphia, Pennsylvania (J.S.M.)
| | - H B Gutstein
- Anesthesiology Institute, Allegheny Health Network, Pittsburgh, Pennsylvania (H.B.G.); University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (S.P., S.R.S., P.T.K.); MD Anderson Cancer Center, Houston, Texas (K.E.B.); and Biostatistics Division, Perelman School of Medicine, Philadelphia, Pennsylvania (J.S.M.)
| |
Collapse
|
11
|
Ashirova E, Contoreggi NH, Johnson MA, Al-Khayat FJ, Calcano GA, Rubin BR, O'Cinneide EM, Zhang Y, Zhou Y, Gregoire L, McEwen BS, Kreek MJ, Milner TA. Oxycodone injections not paired with conditioned place preference have little effect on the hippocampal opioid system in female and male rats. Synapse 2020; 75:e22182. [PMID: 32654187 DOI: 10.1002/syn.22182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/28/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022]
Abstract
Oxycodone (Oxy) conditioned place preference (CPP) in Sprague Dawley rats results in sex-specific alterations in hippocampal opioid circuits in a manner that facilitates opioid-associative learning processes, particularly in females. Here, we examined if Oxy (3 mg/kg, I.P.) or saline (Sal) injections not paired with behavioral testing similarly affect the hippocampal opioid system. Sal-injected females compared to Sal-injected males had: (1) higher densities of cytoplasmic delta opioid receptors (DOR) in GABAergic hilar dendrites suggesting higher baseline reserve DOR pools and (2) elevated phosphorylated DOR levels, but lower phosphorylated mu opioid receptor (MOR) levels in CA3a suggesting that the baseline pools of activated opioid receptors vary in females and males. In contrast to CPP studies, Oxy-injections in the absence of behavioral tests resulted in few changes in the hippocampal opioid system in either females or males. Specifically, Oxy-injected males compared to Sal-injected males had fewer DORs near the plasma membrane of CA3 pyramidal cell dendrites and in CA3 dendritic spines contacted by mossy fibers, and lower pMOR levels in CA3a. Oxy-injected females compared to Sal-injected females had higher total DORs in GABAergic dendrites and lower total MORs in parvalbumin-containing dendrites. Thus, unlike Oxy CPP, Oxy-injections redistributed opioid receptors in hippocampal neurons in a manner that would either decrease (males) or not alter (females) excitability and plasticity processes. These results indicate that the majority of changes within hippocampal opioid circuits that would promote opioid-associative learning processes in both females and males do not occur with Oxy administration alone, and instead must be paired with CPP.
Collapse
Affiliation(s)
- Elina Ashirova
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Megan A Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Fatima J Al-Khayat
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Gabriela A Calcano
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Batsheva R Rubin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Emma M O'Cinneide
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Yong Zhang
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Lennox Gregoire
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
12
|
Sivalingam M, Ogawa S, Parhar IS. Mapping of Morphine-Induced OPRM1 Gene Expression Pattern in the Adult Zebrafish Brain. Front Neuroanat 2020; 14:5. [PMID: 32153369 PMCID: PMC7044135 DOI: 10.3389/fnana.2020.00005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/29/2020] [Indexed: 11/13/2022] Open
Abstract
Morphine is a potent analgesic opiate commonly used in treating pain, and it is also a substance of abuse and highly addictive. Hence, it is vital to discover the action sites of morphine in the brain to increase its efficacy of treatment. In the present study, we aimed at identifying comprehensive neuroanatomical locations that are sensitive to morphine in the adult zebrafish (Danio rerio). We performed in situ hybridization to localize the mu opioid receptor (oprm1) gene and to map the morphine sensitive brain areas using neuronal PAS domain-containing protein 4a (npas4a), an early gene marker. Real-time PCR was used to detect changes in mRNA levels of oprm1 and npas4a in control and acute morphine treated fish (2 mg/L; 20 min). Intense positive oprm1 signals were seen in the telencephalon, preoptic area, habenula, hypothalamic area and periventricular gray zone of the optic tectum. Acute morphine exposure significantly increased oprm1 and npas4a mRNA levels in the medial zone of dorsal telencephalon (Dm), ventral region of the ventral telencephalon (Vv), preoptic area, and in the hypothalamus but a decrease in oprm1 and npas4a signals in the dorsal habenula. This study provides a detailed map of oprm1 localization in the brain, which includes previously unreported oprm1 in the habenula of teleost. Presence of oprm1 in multiple brain sites implies multiple action targets of morphine and potential brain functions which could include reward, cognitive and negative emotions.
Collapse
Affiliation(s)
- Mageswary Sivalingam
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Satoshi Ogawa
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ishwar S Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
13
|
Thakur R, Naik A, Panda A, Raghu P. Regulation of Membrane Turnover by Phosphatidic Acid: Cellular Functions and Disease Implications. Front Cell Dev Biol 2019; 7:83. [PMID: 31231646 PMCID: PMC6559011 DOI: 10.3389/fcell.2019.00083] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/03/2019] [Indexed: 01/23/2023] Open
Abstract
Phosphatidic acid (PA) is a simple glycerophospholipid with a well-established role as an intermediate in phospholipid biosynthesis. In addition to its role in lipid biosynthesis, PA has been proposed to act as a signaling molecule that modulates several aspects of cell biology including membrane transport. PA can be generated in eukaryotic cells by several enzymes whose activity is regulated in the context of signal transduction and enzymes that can metabolize PA thus terminating its signaling activity have also been described. Further, several studies have identified PA binding proteins and changes in their activity are proposed to be mediators of the signaling activity of this lipid. Together these enzymes and proteins constitute a PA signaling toolkit that mediates the signaling functions of PA in cells. Recently, a number of novel genetic models for the analysis of PA function in vivo and analytical methods to quantify PA levels in cells have been developed and promise to enhance our understanding of PA functions. Studies of several elements of the PA signaling toolkit in a single cell type have been performed and are presented to provide a perspective on our understanding of the biochemical and functional organization of pools of PA in a eukaryotic cell. Finally, we also provide a perspective on the potential role of PA in human disease, synthesizing studies from model organisms, human disease genetics and analysis using recently developed PLD inhibitors.
Collapse
Affiliation(s)
- Rajan Thakur
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | - Amruta Naik
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | - Aniruddha Panda
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | - Padinjat Raghu
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| |
Collapse
|
14
|
Temporal dependence of shifts in mu opioid receptor mobility at the cell surface after agonist binding observed by single-particle tracking. Sci Rep 2019; 9:7297. [PMID: 31086197 PMCID: PMC6514008 DOI: 10.1038/s41598-019-43657-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 04/24/2019] [Indexed: 01/13/2023] Open
Abstract
Agonist binding to the mu opioid receptor (MOR) results in conformational changes that allow recruitment of G-proteins, activation of downstream effectors and eventual desensitization and internalization, all of which could affect receptor mobility. The present study employed single particle tracking (SPT) of quantum dot labeled FLAG-tagged MORs to examine shifts in MOR mobility after agonist binding. FLAG-MORs on the plasma membrane were in both mobile and immobile states under basal conditions. Activation of FLAG-MORs with DAMGO caused an acute increase in the fraction of mobile MORs, and free portions of mobile tracks were partially dependent on interactions with G-proteins. In contrast, 10-minute exposure to DAMGO or morphine increased the fraction of immobile FLAG-MORs. While the decrease in mobility with prolonged DAMGO exposure corresponded to an increase in colocalization with clathrin, the increase in colocalization was present in both mobile and immobile FLAG-MORs. Thus, no single mobility state of the receptor accounted for colocalization with clathrin. These findings demonstrate that SPT can be used to track agonist-dependent changes in MOR mobility over time, but that the mobility states observed likely arise from a diverse set of interactions and will be most informative when examined in concert with particular downstream effectors.
Collapse
|
15
|
Reich B, Zhou Y, Goldstein E, Srivats SS, Contoreggi NH, Kogan JF, McEwen BS, Kreek MJ, Milner TA, Gray JD. Chronic immobilization stress primes the hippocampal opioid system for oxycodone-associated learning in female but not male rats. Synapse 2019; 73:e22088. [PMID: 30632204 PMCID: PMC11548942 DOI: 10.1002/syn.22088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/27/2018] [Accepted: 01/06/2019] [Indexed: 12/21/2022]
Abstract
In adult female, but not male, Sprague Dawley rats, chronic immobilization stress (CIS) increases mossy fiber (MF) Leu-Enkephalin levels and redistributes delta- and mu-opioid receptors (DORs and MORs) in hippocampal CA3 pyramidal cells and GABAergic interneurons to promote excitation and learning processes following subsequent opioid exposure. Here, we demonstrate that CIS females, but not males, acquire conditioned place preference (CPP) to oxycodone and that CIS "primes" the hippocampal opioid system in females for oxycodone-associated learning. In CA3b, oxycodone-injected (Oxy) CIS females relative to saline-injected (Sal) CIS females exhibited an increase in the cytoplasmic and total densities of DORs in pyramidal cell dendrites so that they were similar to Sal- and Oxy-CIS males. Consistent with our earlier studies, Sal- and Oxy-CIS females but not CIS males had elevated DOR densities in MF-CA3 dendritic spines, which we have previously shown are important for opioid-mediated long-term potentiation. In the dentate gyrus, Oxy-CIS females had more DOR-labeled interneurons than Sal-CIS females. Moreover, Sal- and Oxy-CIS females compared to both groups of CIS males had elevated levels of DORs and MORs in GABAergic interneuron dendrites, suggesting capacity for greater synthesis or storage of these receptors in circuits important for opioid-mediated disinhibition. However, more plasmalemmal MORs were on large parvalbumin-containing dendrites of Oxy-CIS males compared to Sal-CIS males, suggesting a limited ability for increased granule cell disinhibition. These results suggest that low levels of DORs in MF-CA3 synapses and hilar GABAergic interneurons may contribute to the attenuation of oxycodone CPP in males exposed to CIS.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- CA3 Region, Hippocampal/cytology
- CA3 Region, Hippocampal/drug effects
- CA3 Region, Hippocampal/metabolism
- Conditioning, Classical
- Dendrites/metabolism
- Dentate Gyrus/cytology
- Dentate Gyrus/drug effects
- Dentate Gyrus/metabolism
- Female
- Interneurons/metabolism
- Male
- Oxycodone/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/metabolism
- Repetition Priming
- Restraint, Physical
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- Batsheva Reich
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Ellen Goldstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Sudarshan S. Srivats
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, P.O. Box 24144 - Doha, Qatar
| | - Natalina H. Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Joshua F. Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Jason D. Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| |
Collapse
|
16
|
Ryan JD, Zhou Y, Contoreggi NH, Bshesh FK, Gray JD, Kogan JF, Ben KT, McEwen BS, Jeanne Kreek M, Milner TA. Sex Differences in the Rat Hippocampal Opioid System After Oxycodone Conditioned Place Preference. Neuroscience 2018; 393:236-257. [PMID: 30316908 PMCID: PMC6246823 DOI: 10.1016/j.neuroscience.2018.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022]
Abstract
Although opioid addiction has risen dramatically, the role of gender in addiction has been difficult to elucidate. We previously found sex-dependent differences in the hippocampal opioid system of Sprague-Dawley rats that may promote associative learning relevant to drug abuse. The present studies show that although female and male rats acquired conditioned place preference (CPP) to the mu-opioid receptor (MOR) agonist oxycodone (3 mg/kg, I.P.), hippocampal opioid circuits were differentially altered. In CA3, Leu-Enkephalin-containing mossy fibers had elevated levels in oxycodone CPP (Oxy) males comparable to those in females and sprouted in Oxy-females, suggesting different mechanisms for enhancing opioid sensitivity. Electron microscopy revealed that in Oxy-males delta opioid receptors (DORs) redistributed to mossy fiber-CA3 synapses in a manner resembling females that we previously showed is important for opioid-mediated long-term potentiation. Moreover, in Oxy-females DORs redistributed to CA3 pyramidal cell spines, suggesting the potential for enhanced plasticity processes. In Saline-injected (Sal) females, dentate hilar parvalbumin-containing basket interneuron dendrites had fewer MORs, however plasmalemmal and total MORs increased in Oxy-females. In dentate hilar GABAergic dendrites that contain neuropeptide Y, Sal-females compared to Sal-males had higher plasmalemmal DORs, and near-plasmalemmal DORs increased in Oxy-females. This redistribution of MORs and DORs within hilar interneurons in Oxy-females would potentially enhance disinhibition of granule cells via two different circuits. Together, these results indicate that oxycodone CPP induces sex-dependent redistributions of opioid receptors in hippocampal circuits in a manner facilitating opioid-associative learning processes and may help explain the increased susceptibility of females to opioid addiction acquisition and relapse.
Collapse
Affiliation(s)
- James D Ryan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States.
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Farah K Bshesh
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, P.O. Box 24144 Doha, Qatar
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Joshua F Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Konrad T Ben
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States.
| |
Collapse
|
17
|
Miess E, Gondin AB, Yousuf A, Steinborn R, Mösslein N, Yang Y, Göldner M, Ruland JG, Bünemann M, Krasel C, Christie MJ, Halls ML, Schulz S, Canals M. Multisite phosphorylation is required for sustained interaction with GRKs and arrestins during rapid μ-opioid receptor desensitization. Sci Signal 2018; 11:11/539/eaas9609. [PMID: 30018083 DOI: 10.1126/scisignal.aas9609] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
G protein receptor kinases (GRKs) and β-arrestins are key regulators of μ-opioid receptor (MOR) signaling and trafficking. We have previously shown that high-efficacy opioids such as DAMGO stimulate a GRK2/3-mediated multisite phosphorylation of conserved C-terminal tail serine and threonine residues, which facilitates internalization of the receptor. In contrast, morphine-induced phosphorylation of MOR is limited to Ser375 and is not sufficient to drive substantial receptor internalization. We report how specific multisite phosphorylation controlled the dynamics of GRK and β-arrestin interactions with MOR and show how such phosphorylation mediated receptor desensitization. We showed that GRK2/3 was recruited more quickly than was β-arrestin to a DAMGO-activated MOR. β-Arrestin recruitment required GRK2 activity and MOR phosphorylation, but GRK recruitment also depended on the phosphorylation sites in the C-terminal tail, specifically four serine and threonine residues within the 370TREHPSTANT379 motif. Our results also suggested that other residues outside this motif participated in the initial and transient recruitment of GRK and β-arrestins. We identified two components of high-efficacy agonist desensitization of MOR: a sustained component, which required GRK2-mediated phosphorylation and a potential soluble factor, and a rapid component, which was likely mediated by GRK2 but independent of receptor phosphorylation. Elucidating these complex receptor-effector interactions represents an important step toward a mechanistic understanding of MOR desensitization that leads to the development of tolerance and dependence.
Collapse
Affiliation(s)
- Elke Miess
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia
| | - Arsalan Yousuf
- Discipline of Pharmacology, University of Sydney, New South Wales 2006, Australia
| | - Ralph Steinborn
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany
| | - Nadja Mösslein
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Yunshi Yang
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Martin Göldner
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Julia G Ruland
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Moritz Bünemann
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Cornelius Krasel
- Department of Pharmacology and Toxicology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - MacDonald J Christie
- Discipline of Pharmacology, University of Sydney, New South Wales 2006, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia
| | - Stefan Schulz
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany.
| | - Meritxell Canals
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia.
| |
Collapse
|
18
|
McAlinn HR, Reich B, Contoreggi NH, Kamakura RP, Dyer AG, McEwen BS, Waters EM, Milner TA. Sex Differences in the Subcellular Distribution of Corticotropin-Releasing Factor Receptor 1 in the Rat Hippocampus following Chronic Immobilization Stress. Neuroscience 2018; 383:98-113. [PMID: 29753863 PMCID: PMC5994383 DOI: 10.1016/j.neuroscience.2018.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/01/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Abstract
Corticotropin-releasing factor receptors (CRFR1) contribute to stress-induced adaptations in hippocampal structure and function that can affect learning and memory processes. Our prior studies showed that female rats with elevated estrogens compared to males have more plasmalemmal CRFR1 in CA1 pyramidal cells, suggesting a greater sensitivity to stress. Here, we examined the distribution of hippocampal CRFR1 following chronic immobilization stress (CIS) in female and male rats using immuno-electron microscopy. Without stress, total CRFR1 dendritic levels were higher in females in CA1 and in males in the hilus; moreover, plasmalemmal CRFR1 was elevated in pyramidal cell dendrites in CA1 in females and in CA3 in males. Following CIS, near-plasmalemmal CRFR1 increased in CA1 pyramidal cell dendrites in males but not to levels of control or CIS females. In CA3 and the hilus, CIS decreased cytoplasmic and total CRFR1 in dendrites in males only. These results suggest that in naive rats, CRF could induce a greater activation of CA1 pyramidal cells in females than males. Moreover, after CIS, which leads to even greater sex differences in CRFR1 by trafficking it to different subcellular compartments, CRF could enhance activation of CA1 pyramidal cells in males but to a lesser extent than either unstressed or CIS females. Additionally, CA3 pyramidal cells and inhibitory interneurons in males have heightened sensitivity to CRF, regardless of stress state. These sex differences in CRFR1 distribution and trafficking in the hippocampus may contribute to reported sex differences in hippocampus-dependent learning processes in baseline conditions and following chronic stress.
Collapse
Affiliation(s)
- Helena R McAlinn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Batsheva Reich
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Andreina G Dyer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
19
|
Bao F, Li CL, Chen XQ, Lu YJ, Bao L, Zhang X. Clinical opioids differentially induce co-internalization of μ- and δ-opioid receptors. Mol Pain 2018; 14:1744806918769492. [PMID: 29587571 PMCID: PMC5898661 DOI: 10.1177/1744806918769492] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Opioid receptors play an important role in mediating the spinal analgesia. The μ-opioid receptor is the major target of opioid drugs widely used in clinics. However, the regulatory mechanisms of analgesic effect and tolerance for clinical μ-opioid receptor-targeting opioids remain to be fully investigated. Previous studies showed the interaction of δ-opioid receptor with μ-opioid receptor to form the μ-opioid receptor/δ-opioid receptor heteromers that could be processed in the degradation pathway after δ-opioid receptor agonist treatment. Here, we showed that clinical μ-opioid receptor-targeting opioids, morphine, fentanyl, and methadone, but not tramadol, caused μ-opioid receptor co-internalization with δ-opioid receptors in both transfected human embryonic kidney 293 cells and primary sensory neurons. Prolonged treatment of morphine led to μ-opioid receptor co-degradation with δ-opioid receptors. Furthermore, fentanyl and methadone, but not tramadol, induced the drug tolerance similar to morphine. Thus, the clinical μ-opioid receptor-targeting opioids including morphine, fentanyl, and methadone induce μ-opioid receptor co-internalization with δ-opioid receptors, which may be involved in the analgesic tolerance of these opioids.
Collapse
Affiliation(s)
- Fenghua Bao
- 1 Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,2 University of Chinese Academy of Sciences, Shanghai, China
| | - Chang-Lin Li
- 1 Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,2 University of Chinese Academy of Sciences, Shanghai, China.,3 Shanghai Clinical Research Center, Chinese Academy of Sciences/XuHui Central Hospital, Shanghai, China
| | - Xu-Qiao Chen
- 4 State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Ying-Jin Lu
- 1 Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,2 University of Chinese Academy of Sciences, Shanghai, China.,3 Shanghai Clinical Research Center, Chinese Academy of Sciences/XuHui Central Hospital, Shanghai, China
| | - Lan Bao
- 4 State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.,5 School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xu Zhang
- 1 Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,2 University of Chinese Academy of Sciences, Shanghai, China.,5 School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
20
|
Margolis EB, Fujita W, Devi LA, Fields HL. Two delta opioid receptor subtypes are functional in single ventral tegmental area neurons, and can interact with the mu opioid receptor. Neuropharmacology 2017. [PMID: 28645621 DOI: 10.1016/j.neuropharm.2017.06.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The mu and delta opioid receptors (MOR and DOR) are highly homologous members of the opioid family of GPCRs. There is evidence that MOR and DOR interact, however the extent to which these interactions occur in vivo and affect synaptic function is unknown. There are two stable DOR subtypes: DPDPE sensitive (DOR1) and deltorphin II sensitive (DOR2); both agonists are blocked by DOR selective antagonists. Robust motivational effects are produced by local actions of both MOR and DOR ligands in the ventral tegmental area (VTA). Here we demonstrate that a majority of both dopaminergic and non-dopaminergic VTA neurons express combinations of functional DOR1, DOR2, and/or MOR, and that within a single VTA neuron, DOR1, DOR2, and MOR agonists can differentially couple to downstream signaling pathways. As reported for the MOR agonist DAMGO, DPDPE and deltorphin II produced either a predominant K+ dependent hyperpolarization or a Cav2.1 mediated depolarization in different neurons. In some neurons DPDPE and deltorphin II produced opposite responses. Excitation, inhibition, or no effect by DAMGO did not predict the response to DPDPE or deltorphin II, arguing against a MOR-DOR interaction generating DOR subtypes. However, in a subset of VTA neurons the DOR antagonist TIPP-Ψ augmented DAMGO responses; we also observed DPDPE or deltorphin II responses augmented by the MOR selective antagonist CTAP. These findings directly support the existence of two independent, stable forms of the DOR, and show that MOR and DOR can interact in some neurons to alter downstream signaling.
Collapse
Affiliation(s)
- Elyssa B Margolis
- Department of Neurology, The Wheeler Center for the Neurobiology of Addiction, Alcoholism and Addiction Research Group, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Wakako Fujita
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Howard L Fields
- Department of Neurology, The Wheeler Center for the Neurobiology of Addiction, Alcoholism and Addiction Research Group, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
21
|
Mazid S, Hall BS, Odell SC, Stafford K, Dyer AD, Van Kempen TA, Selegean J, McEwen BS, Waters EM, Milner TA. Sex differences in subcellular distribution of delta opioid receptors in the rat hippocampus in response to acute and chronic stress. Neurobiol Stress 2016; 5:37-53. [PMID: 27981195 PMCID: PMC5145913 DOI: 10.1016/j.ynstr.2016.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 10/18/2016] [Accepted: 11/07/2016] [Indexed: 02/06/2023] Open
Abstract
Drug addiction requires associative learning processes that critically involve hippocampal circuits, including the opioid system. We recently found that acute and chronic stress, important regulators of addictive processes, affect hippocampal opioid levels and mu opioid receptor trafficking in a sexually dimorphic manner. Here, we examined whether acute and chronic stress similarly alters the levels and trafficking of hippocampal delta opioid receptors (DORs). Immediately after acute immobilization stress (AIS) or one-day after chronic immobilization stress (CIS), the brains of adult female and male rats were perfusion-fixed with aldehydes. The CA3b region and the dentate hilus of the dorsal hippocampus were quantitatively analyzed by light microscopy using DOR immunoperoxidase or dual label electron microscopy for DOR using silver intensified immunogold particles (SIG) and GABA using immunoperoxidase. At baseline, females compared to males had more DORs near the plasmalemma of pyramidal cell dendrites and about 3 times more DOR-labeled CA3 dendritic spines contacted by mossy fibers. In AIS females, near-plasmalemmal DOR-SIGs decreased in GABAergic hilar dendrites. However, in AIS males, near-plasmalemmal DOR-SIGs increased in CA3 pyramidal cell and hilar GABAergic dendrites and the percentage of CA3 dendritic spines contacted by mossy fibers increased to about half that seen in unstressed females. Conversely, after CIS, near-plasmalemmal DOR-SIGs increased in hilar GABA-labeled dendrites of females whereas in males plasmalemmal DOR-SIGs decreased in CA3 pyramidal cell dendrites and near-plasmalemmal DOR-SIGs decreased hilar GABA-labeled dendrites. As CIS in females, but not males, redistributed DOR-SIGs near the plasmalemmal of hilar GABAergic dendrites, a subsequent experiment examined the acute affect of oxycodone on the redistribution of DOR-SIGs in a separate cohort of CIS females. Plasmalemmal DOR-SIGs were significantly elevated on hilar interneuron dendrites one-hour after oxycodone (3 mg/kg, I.P.) administration compared to saline administration in CIS females. These data indicate that DORs redistribute within CA3 pyramidal cells and dentate hilar GABAergic interneurons in a sexually dimorphic manner that would promote activation and drug related learning in males after AIS and in females after CIS. Females have more near-plasmalemmal DORs in pyramidal CA3 dendrites than males. Acute stress in males relocates DORs in CA3 & GABA dendrites to promote activation. Chronic stress in females relocates DORs in GABA dendrites in females to promote activation. Chronic stress in males relocates DORs in GABA dendrites opposite of females. DOR-stress relocation may contribute to sexually dimorphic effects on drug related learning.
Collapse
Affiliation(s)
- Sanoara Mazid
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Baila S Hall
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States
| | - Shannon C Odell
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States
| | - Khalifa Stafford
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Andreina D Dyer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Tracey A Van Kempen
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States
| | - Jane Selegean
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, United States; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States
| |
Collapse
|
22
|
Halls ML, Yeatman HR, Nowell CJ, Thompson GL, Gondin AB, Civciristov S, Bunnett NW, Lambert NA, Poole DP, Canals M. Plasma membrane localization of the μ-opioid receptor controls spatiotemporal signaling. Sci Signal 2016; 9:ra16. [PMID: 26861044 DOI: 10.1126/scisignal.aac9177] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Differential regulation of the μ-opioid receptor (MOR), a G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor, contributes to the clinically limiting effects of opioid analgesics, such as morphine. We used biophysical approaches to quantify spatiotemporal MOR signaling in response to different ligands. In human embryonic kidney (HEK) 293 cells overexpressing MOR, morphine caused a Gβγ-dependent increase in plasma membrane-localized protein kinase C (PKC) activity, which resulted in a restricted distribution of MOR within the plasma membrane and induced sustained cytosolic extracellular signal-regulated kinase (ERK) signaling. In contrast, the synthetic opioid peptide DAMGO ([d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin) enabled receptor redistribution within the plasma membrane, resulting in transient increases in cytosolic and nuclear ERK activity, and, subsequently, receptor internalization. When Gβγ subunits or PKCα activity was inhibited or when the carboxyl-terminal phosphorylation sites of MOR were mutated, morphine-activated MOR was released from its restricted plasma membrane localization and stimulated a transient increase in cytosolic and nuclear ERK activity in the absence of receptor internalization. Thus, these data suggest that the ligand-induced redistribution of MOR within the plasma membrane, and not its internalization, controls its spatiotemporal signaling.
Collapse
Affiliation(s)
- Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Holly R Yeatman
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Cameron J Nowell
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Georgina L Thompson
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Arisbel Batista Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Srgjan Civciristov
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Nigel W Bunnett
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. Department of Anesthesia and Perioperative Medicine, Monash University, Melbourne, Victoria 3004, Australia. Department of Pharmacology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Nevin A Lambert
- Department of Toxicology and Pharmacology, Georgia Regents University, Augusta, GA 30912, USA
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Meritxell Canals
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| |
Collapse
|
23
|
Hahn YK, Paris JJ, Lichtman AH, Hauser KF, Sim-Selley LJ, Selley DE, Knapp PE. Central HIV-1 Tat exposure elevates anxiety and fear conditioned responses of male mice concurrent with altered mu-opioid receptor-mediated G-protein activation and β-arrestin 2 activity in the forebrain. Neurobiol Dis 2016; 92:124-36. [PMID: 26845176 DOI: 10.1016/j.nbd.2016.01.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/31/2015] [Accepted: 01/19/2016] [Indexed: 02/06/2023] Open
Abstract
Co-exposure to opiates and HIV/HIV proteins results in enhanced CNS morphological and behavioral deficits in HIV(+) individuals and in animal models. Opiates with abuse liability, such as heroin and morphine, bind preferentially to and have pharmacological actions through μ-opioid-receptors (MORs). The mechanisms underlying opiate-HIV interactions are not understood. Exposure to the HIV-1 transactivator of transcription (Tat) protein causes neurodegenerative outcomes that parallel many aspects of the human disease. We have also observed that in vivo exposure to Tat results in apparent changes in morphine efficacy, and thus have hypothesized that HIV proteins might alter MOR activation. To test our hypothesis, MOR-mediated G-protein activation was determined in neuroAIDS-relevant forebrain regions of transgenic mice with inducible CNS expression of HIV-1 Tat. G-protein activation was assessed by MOR agonist-stimulated [(35)S]guanosine-5'-O-(3-thio)triphosphate ([(35)S]GTPγS) autoradiography in brain sections, and in concentration-effect curves of MOR agonist-stimulated [(35)S]GTPγS binding in membranes isolated from specific brain regions. Comparative studies were done using the MOR-selective agonist DAMGO ([D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin) and a more clinically relevant agonist, morphine. Tat exposure reduced MOR-mediated G-protein activation in an agonist, time, and regionally dependent manner. Levels of the GPCR regulatory protein β-arrestin-2, which is involved in MOR desensitization, were found to be elevated in only one affected brain region, the amygdala; amygdalar β-arrestin-2 also showed a significantly increased association with MOR by co-immunoprecipitation, suggesting decreased availability of MOR. Interestingly, this correlated with changes in anxiety and fear-conditioned extinction, behaviors that have substantial amygdalar input. We propose that HIV-1 Tat alters the intrinsic capacity of MOR to signal in response to agonist binding, possibly via a mechanism involving altered expression and/or function of β-arrestin-2.
Collapse
Affiliation(s)
- Yun K Hahn
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA
| | - Jason J Paris
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Kurt F Hauser
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA; Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Laura J Sim-Selley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Dana E Selley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Pamela E Knapp
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA; Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA.
| |
Collapse
|
24
|
Sex differences in NMDA GluN1 plasticity in rostral ventrolateral medulla neurons containing corticotropin-releasing factor type 1 receptor following slow-pressor angiotensin II hypertension. Neuroscience 2015; 307:83-97. [PMID: 26306872 DOI: 10.1016/j.neuroscience.2015.08.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/12/2015] [Accepted: 08/14/2015] [Indexed: 11/15/2022]
Abstract
There are profound, yet incompletely understood, sex differences in the neurogenic regulation of blood pressure. Both corticotropin signaling and glutamate receptor plasticity, which differ between males and females, are known to play important roles in the neural regulation of blood pressure. However, the relationship between hypertension and glutamate plasticity in corticotropin-releasing factor (CRF)-receptive neurons in brain cardiovascular regulatory areas, including the rostral ventrolateral medulla (RVLM) and paraventricular nucleus of the hypothalamus (PVN), is not understood. In the present study, we used dual-label immuno-electron microscopy to analyze sex differences in slow-pressor angiotensin II (AngII) hypertension with respect to the subcellular distribution of the obligatory NMDA glutamate receptor subunit 1 (GluN1) subunit of the N-methyl-D-aspartate receptor (NMDAR) in the RVLM and PVN. Studies were conducted in mice expressing the enhanced green fluorescence protein (EGFP) under the control of the CRF type 1 receptor (CRF1) promoter (i.e., CRF1-EGFP reporter mice). By light microscopy, GluN1-immunoreactivity (ir) was found in CRF1-EGFP neurons of the RVLM and PVN. Moreover, in both regions tyrosine hydroxylase (TH) was found in CRF1-EGFP neurons. In response to AngII, male mice showed an elevation in blood pressure that was associated with an increase in the proportion of GluN1 on presumably functional areas of the plasma membrane (PM) in CRF1-EGFP dendritic profiles in the RVLM. In female mice, AngII was neither associated with an increase in blood pressure nor an increase in PM GluN1 in the RVLM. Unlike the RVLM, AngII-mediated hypertension had no effect on GluN1 localization in CRF1-EGFP dendrites in the PVN of either male or female mice. These studies provide an anatomical mechanism for sex-differences in the convergent modulation of RVLM catecholaminergic neurons by CRF and glutamate. Moreover, these results suggest that sexual dimorphism in AngII-induced hypertension is reflected by NMDA receptor trafficking in presumptive sympathoexcitatory neurons in the RVLM.
Collapse
|
25
|
Allouche S, Noble F, Marie N. Opioid receptor desensitization: mechanisms and its link to tolerance. Front Pharmacol 2014; 5:280. [PMID: 25566076 PMCID: PMC4270172 DOI: 10.3389/fphar.2014.00280] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/02/2014] [Indexed: 02/04/2023] Open
Abstract
Opioid receptors (OR) are part of the class A of G-protein coupled receptors and the target of the opiates, the most powerful analgesic molecules used in clinic. During a protracted use, a tolerance to analgesic effect develops resulting in a reduction of the effectiveness. So understanding mechanisms of tolerance is a great challenge and may help to find new strategies to tackle this side effect. This review will summarize receptor-related mechanisms that could underlie tolerance especially receptor desensitization. We will focus on the latest data obtained on molecular mechanisms involved in opioid receptor desensitization: phosphorylation, receptor uncoupling, internalization, and post-endocytic fate of the receptor.
Collapse
Affiliation(s)
- Stéphane Allouche
- Laboratoire de Signalisation, Électrophysiologie et Imagerie des Lésions D'ischémie-Reperfusion Myocardique, Université de Caen, UPRES EA 4650, IFR 146 ICORE Caen, France
| | - Florence Noble
- Centre National de la Recherche Scientifique, ERL 3649 Paris, France ; Institut National de la Santé et de la Recherche Médicale, UMR-S 1124 Paris, France ; Université Paris Descartes, Neuroplasticité et Thérapies des Addictions Paris, France
| | - Nicolas Marie
- Centre National de la Recherche Scientifique, ERL 3649 Paris, France ; Institut National de la Santé et de la Recherche Médicale, UMR-S 1124 Paris, France ; Université Paris Descartes, Neuroplasticité et Thérapies des Addictions Paris, France
| |
Collapse
|
26
|
Glück L, Loktev A, Moulédous L, Mollereau C, Law PY, Schulz S. Loss of morphine reward and dependence in mice lacking G protein-coupled receptor kinase 5. Biol Psychiatry 2014; 76:767-74. [PMID: 24629717 PMCID: PMC4119866 DOI: 10.1016/j.biopsych.2014.01.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 01/28/2014] [Accepted: 01/28/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND The clinical benefits of opioid drugs are counteracted by the development of tolerance and addiction. We provide in vivo evidence for the involvement of G protein-coupled receptor kinases (GRKs) in opioid dependence in addition to their roles in agonist-selective mu-opioid receptor (MOR) phosphorylation. METHODS In vivo MOR phosphorylation was examined by immunoprecipitation and nanoflow liquid chromatography-tandem mass spectrometry analysis. Using the hot-plate and conditioned place preference test, we investigated opioid-related antinociception and reward effects in mice lacking GRK3 or GRK5. RESULTS Etonitazene and fentanyl stimulated the in vivo phosphorylation of multiple carboxyl-terminal phosphate acceptor sites, including threonine 370, serine 375, and threonine 379, which was predominantly mediated by GRK3. By contrast, morphine promoted a selective phosphorylation of serine 375 that was predominantly mediated by GRK5. In contrast to GRK3 knockout mice, GRK5 knockout mice exhibited reduced antinociceptive responses after morphine administration and developed morphine tolerance similar to wild-type mice but fewer signs of physical dependence. Also, morphine was ineffective in inducing conditioned place preference in GRK5 knockout mice, whereas cocaine conditioned place preference was retained. However, the reward properties of morphine were evident in knock-in mice expressing a phosphorylation-deficient S375A mutation of the MOR. CONCLUSIONS These findings show for the first time that MOR phosphorylation is regulated by agonist-selective recruitment of distinct GRK isoforms that influence different opioid-related behaviors. Modulation of GRK5 function could serve as a new approach for preventing addiction to opioids, while maintaining the analgesic properties of opioid drugs at an effective level.
Collapse
Affiliation(s)
- Laura Glück
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, Germany
| | - Anastasia Loktev
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, Germany
| | - Lionel Moulédous
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | - Catherine Mollereau
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse, UMR 5089, 205 Route de Narbonne, 31077 Toulouse Cedex, France
| | - Ping-Yee Law
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, 55455 USA
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
27
|
Kelly E. Efficacy and ligand bias at the μ-opioid receptor. Br J Pharmacol 2014; 169:1430-46. [PMID: 23646826 DOI: 10.1111/bph.12222] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 04/10/2013] [Accepted: 04/20/2013] [Indexed: 12/11/2022] Open
Abstract
In order to describe drug action at a GPCR, a full understanding of the pharmacological terms affinity, efficacy and potency is necessary. This is true whether comparing the ability of different agonists to produce a measurable response in a cell or tissue, or determining the relative ability of an agonist to activate a single receptor subtype and produce multiple responses. There is a great deal of interest in the μ-opioid receptor (MOP receptor) and the ligands that act at this GPCR not only because of the clinically important analgesic effects produced by MOP agonists but also because of their liability to induce adverse effects such as respiratory depression and dependence. Our understanding of the mechanisms underlying these effects, as well as the ability to develop new, more effective MOP receptor drugs, depends upon the accurate determination of the efficacy with which these ligands induce coupling of MOP receptors to downstream signalling events. In this review, which is written with the minimum of mathematical content, the basic meaning of terms including efficacy, intrinsic activity and intrinsic efficacy is discussed, along with their relevance to the field of MOP receptor pharmacology, and in particular in relation to biased agonism at this important GPCR.
Collapse
Affiliation(s)
- E Kelly
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK.
| |
Collapse
|
28
|
Massotte D. In vivo opioid receptor heteromerization: where do we stand? Br J Pharmacol 2014; 172:420-34. [PMID: 24666391 DOI: 10.1111/bph.12702] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/17/2014] [Accepted: 03/19/2014] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Opioid receptors are highly homologous GPCRs that modulate brain function at all levels of neural integration, including autonomous, sensory, emotional and cognitive processing. Opioid receptors functionally interact in vivo, but the underlying mechanisms involving direct receptor-receptor interactions, affecting signalling pathways or engaging different neuronal circuits, remain unsolved. Heteromer formation through direct physical interaction between two opioid receptors or between an opioid receptor and a non-opioid one has been postulated and can be characterized by specific ligand binding, receptor signalling and trafficking properties. However, despite numerous studies in heterologous systems, evidence for physical proximity in vivo is only available for a limited number of opioid heteromers, and their physiopathological implication remains largely unknown mostly due to the lack of appropriate tools. Nonetheless, data collected so far using endogenous receptors point to a crucial role for opioid heteromers as a molecular entity that could underlie human pathologies such as alcoholism, acute or chronic pain as well as psychiatric disorders. Opioid heteromers therefore stand as new therapeutic targets for the drug discovery field. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- D Massotte
- Institut des Neurosciences Cellulaires et Intégratives, INCI, Strasbourg, France
| |
Collapse
|
29
|
Jaremko KM, Thompson NL, Reyes BAS, Jin J, Ebersole B, Jenney CB, Grigson PS, Levenson R, Berrettini WH, Van Bockstaele EJ. Morphine-induced trafficking of a mu-opioid receptor interacting protein in rat locus coeruleus neurons. Prog Neuropsychopharmacol Biol Psychiatry 2014; 50:53-65. [PMID: 24333843 PMCID: PMC3928604 DOI: 10.1016/j.pnpbp.2013.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/26/2013] [Accepted: 12/05/2013] [Indexed: 12/14/2022]
Abstract
Opiate addiction is a devastating health problem, with approximately 2million people currently addicted to heroin or non-medical prescription opiates in the United States alone. In neurons, adaptations in cell signaling cascades develop following opioid actions at the mu opioid receptor (MOR). A novel putative target for intervention involves interacting proteins that may regulate trafficking of MOR. Morphine has been shown to induce a re-distribution of a MOR-interacting protein Wntless (WLS, a transport molecule necessary for secretion of neurotrophic Wnt proteins), from cytoplasmic to membrane compartments in rat striatal neurons. Given its opiate-sensitivity and its well-characterized molecular and cellular adaptations to morphine exposure, we investigated the anatomical distribution of WLS and MOR in the rat locus coeruleus (LC)-norepinephrine (NE) system. Dual immunofluorescence microscopy was used to test the hypothesis that WLS is localized to noradrenergic neurons of the LC and that WLS and MOR co-exist in common LC somatodendritic processes, providing an anatomical substrate for their putative interactions. We also hypothesized that morphine would influence WLS distribution in the LC. Rats received saline, morphine or the opiate agonist [d-Ala2, N-Me-Phe4, Gly-ol5]-enkephalin (DAMGO), and tissue sections through the LC were processed for immunogold-silver detection of WLS and MOR. Statistical analysis showed a significant re-distribution of WLS to the plasma membrane following morphine treatment in addition to an increase in the proximity of gold-silver labels for MOR and WLS. Following DAMGO treatment, MOR and WLS were predominantly localized within the cytoplasmic compartment when compared to morphine and control. In a separate cohort of rats, brains were obtained from saline-treated or heroin self-administering male rats for pulldown co-immunoprecipitation studies. Results showed an increased association of WLS and MOR following heroin exposure. As the LC-NE system is important for cognition as well as decisions underlying substance abuse, adaptations in WLS trafficking and expression may play a role in modulating MOR function in the LC and contribute to the negative sequelae of opiate exposure on executive function.
Collapse
Affiliation(s)
- Kellie M Jaremko
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Nicholas L Thompson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Beverly A S Reyes
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States.
| | - Jay Jin
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Brittany Ebersole
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Christopher B Jenney
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Patricia S Grigson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Robert Levenson
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Wade H Berrettini
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, United States
| | - Elisabeth J Van Bockstaele
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| |
Collapse
|
30
|
Niikura K, Ho A, Kreek MJ, Zhang Y. Oxycodone-induced conditioned place preference and sensitization of locomotor activity in adolescent and adult mice. Pharmacol Biochem Behav 2013; 110:112-6. [PMID: 23827650 DOI: 10.1016/j.pbb.2013.06.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/25/2013] [Accepted: 06/22/2013] [Indexed: 10/26/2022]
Abstract
Nonmedical use of the prescription opioid oxycodone has become a major public health problem in the United States, with special concern for adolescents. Although adults and adolescents have different sensitivities for drugs, little is known about the rewarding effects of oxycodone in adolescents compared to adults, even in rodent models. Here, we investigate sensitivity to oxycodone by the conditioned place preference assay of conditioned reward, and effect on the locomotor activity in adolescent (4 weeks old) and adult (10 weeks old) C57BL/6J mice. Mice of both ages were trained with multiple doses of oxycodone (0, 0.3, 1, and 3 mg/kg) and showed conditioned preference in a dose-dependent manner. The adult mice developed conditioned preference to the lowest dose tested (0.3 mg/kg), but adolescent mice did not. Dose-dependent oxycodone-induced increases in locomotor activity were observed across the conditioning session. Interestingly, adolescent mice developed greater sensitization to the locomotor-activating effects of oxycodone than adult mice. Thus differences in sensitivity to oxycodone, such as the lower initial sensitivity for conditioned preference but greater locomotor sensitization in adolescent mice, may indicate contributing factors in oxycodone abuse and later addiction in human adolescents.
Collapse
Affiliation(s)
- Keiichi Niikura
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | | | | | | |
Collapse
|
31
|
Andreoni A, Nardo L, Bondani M, Zhao B, Roberts JE. Time-resolved fluorescence studies of fullerene derivatives. J Phys Chem B 2013; 117:7203-9. [PMID: 23646878 DOI: 10.1021/jp400877h] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fullerene (nano-C60) and its water-soluble derivatives have several clinical applications including use as a drug carrier to bypass the blood-ocular and blood-brain barriers. However, in vitro and in vivo detection of these nanomaterials is limited by their very low fluorescence quantum yield. The accumulation of fullerene and its derivatives in cells is particularly difficult to measure using standard fluorescence microscopy because their fluorescence is barely detectable in aqueous media. We have developed a time-correlated single-photon counting apparatus with which we were not only able to detect the fluorescence of fullerene and its derivatives in water but could also measure fluorescence temporal decays and determine lifetimes in the range of tens of picoseconds. The compounds studied in this report are C60 (fullerene), the partially hydrogenated hydride C60H36, a monomeric cyclodextrin complexed fullerene [(γ-CyD)2/C60], and C60(OH)24 (fullerol). In addition, we examined the effect of aggregation on photophysical properties and identified a very short lifetime component belonging to the fluorescence decay of monomeric fullerene, which is lost with increasing aggregation. These data will help to design nanoparticles that have the appropriate structural and photophysical properties to ultimately be of use in a clinical setting.
Collapse
Affiliation(s)
- Alessandra Andreoni
- Department of Science and High Tech and C.N.I.S.M., Unita' di Como, University of Insubria, Como, 22100 Italy.
| | | | | | | | | |
Collapse
|
32
|
Desai SJ, Upadhya MA, Subhedar NK, Kokare DM. NPY mediates reward activity of morphine, via NPY Y1 receptors, in the nucleus accumbens shell. Behav Brain Res 2013; 247:79-91. [DOI: 10.1016/j.bbr.2013.03.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 03/05/2013] [Accepted: 03/09/2013] [Indexed: 10/27/2022]
|
33
|
Williams JT, Ingram SL, Henderson G, Chavkin C, von Zastrow M, Schulz S, Koch T, Evans CJ, Christie MJ. Regulation of μ-opioid receptors: desensitization, phosphorylation, internalization, and tolerance. Pharmacol Rev 2013; 65:223-54. [PMID: 23321159 PMCID: PMC3565916 DOI: 10.1124/pr.112.005942] [Citation(s) in RCA: 619] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Morphine and related µ-opioid receptor (MOR) agonists remain among the most effective drugs known for acute relief of severe pain. A major problem in treating painful conditions is that tolerance limits the long-term utility of opioid agonists. Considerable effort has been expended on developing an understanding of the molecular and cellular processes that underlie acute MOR signaling, short-term receptor regulation, and the progression of events that lead to tolerance for different MOR agonists. Although great progress has been made in the past decade, many points of contention and controversy cloud the realization of this progress. This review attempts to clarify some confusion by clearly defining terms, such as desensitization and tolerance, and addressing optimal pharmacological analyses for discerning relative importance of these cellular mechanisms. Cellular and molecular mechanisms regulating MOR function by phosphorylation relative to receptor desensitization and endocytosis are comprehensively reviewed, with an emphasis on agonist-biased regulation and areas where knowledge is lacking or controversial. The implications of these mechanisms for understanding the substantial contribution of MOR signaling to opioid tolerance are then considered in detail. While some functional MOR regulatory mechanisms contributing to tolerance are clearly understood, there are large gaps in understanding the molecular processes responsible for loss of MOR function after chronic exposure to opioids. Further elucidation of the cellular mechanisms that are regulated by opioids will be necessary for the successful development of MOR-based approaches to new pain therapeutics that limit the development of tolerance.
Collapse
Affiliation(s)
- John T Williams
- Vollum Institute, Oregon Health Sciences University, Portland, Oregon, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
von Zastrow M, Williams JT. Modulating neuromodulation by receptor membrane traffic in the endocytic pathway. Neuron 2012; 76:22-32. [PMID: 23040804 DOI: 10.1016/j.neuron.2012.09.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cellular responsiveness to many neuromodulators is controlled by endocytosis of the transmembrane receptors that transduce their effects. Endocytic membrane trafficking of particular neuromodulator receptors exhibits remarkable diversity and specificity, determined largely by molecular sorting operations that guide receptors at trafficking branchpoints after endocytosis. In this Review, we discuss recent progress in elucidating mechanisms mediating the molecular sorting of neuromodulator receptors in the endocytic pathway. There is emerging evidence that endocytic trafficking of neuromodulator receptors, in addition to influencing longer-term cellular responsiveness under conditions of prolonged or repeated activation, may also affect the acute response. Physiological and pathological consequences of defined receptor trafficking events are only now being elucidated, but it is already apparent that endocytosis of neuromodulator receptors has a significant impact on the actions of therapeutic drugs. The present data also suggest, conversely, that mechanisms of receptor endocytosis and molecular sorting may themselves represent promising targets for therapeutic manipulation.
Collapse
Affiliation(s)
- Mark von Zastrow
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA 94158, USA.
| | | |
Collapse
|
35
|
Dang VC, Christie MJ. Mechanisms of rapid opioid receptor desensitization, resensitization and tolerance in brain neurons. Br J Pharmacol 2012; 165:1704-1716. [PMID: 21564086 DOI: 10.1111/j.1476-5381.2011.01482.x] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Agonists acting on µ-opioid receptors (MOR) are very effective analgesics but cause tolerance during long-term or repeated exposure. Intensive efforts have been made to find novel opioid agonists that are efficacious analgesics but can elude the signalling events that cause tolerance. µ-Opioid agonists differentially couple to downstream signalling mechanisms. Some agonists, such as enkephalins, D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO), methadone and sufentanyl are efficacious at mediating G-protein and effector coupling, as well as triggering MOR regulatory events that include MOR phosphorylation, β-arrestin binding, receptor endocytosis and recycling. By contrast, morphine and closely related alkaloids can mediate efficacious MOR-effector coupling but poorly trigger receptor regulation. Several models have been proposed to relate differential MOR regulation by different opioids with their propensity to cause tolerance. Most are based on dogma that β-arrestin-2 (βarr-2) binding causes MOR desensitization and/or that MOR endocytosis and recycling are required for receptor resensitization. This review will examine some of these notions in light of recent evidence establishing that MOR dephosphorylation and resensitization do not require endocytosis. Recent evidence from opioid-treated animals also suggests that impaired MOR-effector coupling is driven, at least in part, by enhanced desensitization, as well as impaired resensitization that appears to be βarr-2 dependent. Better understanding of how chronic exposure to opioids alters receptor regulatory mechanisms may facilitate the development of effective analgesics that produce limited tolerance.
Collapse
Affiliation(s)
- Vu C Dang
- Department of Psychiatry, University of California, San Francisco, CA, USABrain & Mind Research Institute, University of Sydney, NSW, Australia
| | - MacDonald J Christie
- Department of Psychiatry, University of California, San Francisco, CA, USABrain & Mind Research Institute, University of Sydney, NSW, Australia
| |
Collapse
|
36
|
Whistler JL. Examining the role of mu opioid receptor endocytosis in the beneficial and side-effects of prolonged opioid use: from a symposium on new concepts in mu-opioid pharmacology. Drug Alcohol Depend 2012; 121:189-204. [PMID: 22226706 PMCID: PMC4224378 DOI: 10.1016/j.drugalcdep.2011.10.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 10/20/2011] [Accepted: 10/22/2011] [Indexed: 12/31/2022]
Abstract
Opioid drugs remain the gold standard for the treatment of severe pain, both acute/post-surgical and chronic. However, the utility of opioid drugs for the treatment of chronic pain is compromised by the development of analgesic tolerance which, in turn, leads to dose-escalation and increased likelihood of dangerous side effects, including dependence. Consequently, there remains resistance among clinicians and the general population to using opiates for pain management because of risk of "addiction." These fears are not unwarranted. More than 2.5 million people begin abusing opioid painkillers each year, and prescription opioid abuse is now the second most common type of illegal drug use after marijuana. Some abusers become dependent due to recreational use of prescription painkillers. However, many abusers are among the 40 million people suffering from chronic pain, and developed dependence while using the drugs for legitimate purposes. Both of these trends highlight the need to develop opioid therapeutics with a reduced liability to cause tolerance, dependence and addiction. Identifying the ideal properties of opioid drugs that would retain analgesia but reduce these side-effects has been a goal of my laboratory for more than a decade. During this time, we have proposed the novel hypothesis that opioid drugs that promote desensitization, endocytosis and recycling of the mu-opioid-receptor (MOR) will retain analgesic efficacy, but will have a reduced liability to cause tolerance, dependence and addiction. We have generated substantial data, both pharmacological and genetic to suggest that our hypothesis is a valid one. These data are summarized in this review.
Collapse
Affiliation(s)
- Jennifer L. Whistler
- Department of Neurology, Ernest Gallo Clinic and Research Center, University of California, San Francisco, 5858 Horton St. Suite 200, Emeryville, CA 94608, tel: 510 985-3127, fax: 510 985-3101,
| |
Collapse
|
37
|
Lau EK, Trester-Zedlitz M, Trinidad JC, Kotowski SJ, Krutchinsky AN, Burlingame AL, von Zastrow M. Quantitative encoding of the effect of a partial agonist on individual opioid receptors by multisite phosphorylation and threshold detection. Sci Signal 2011; 4:ra52. [PMID: 21868358 DOI: 10.1126/scisignal.2001748] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In comparison to endogenous ligands of seven-transmembrane receptors, which typically act as full agonists, many drugs act as partial agonists. Partial agonism is best described as a "macroscopic" property that is manifest at the level of physiological systems or cell populations; however, whether partial agonists also encode discrete regulatory information at the "microscopic" level of individual receptors is not known. Here, we addressed this question by focusing on morphine, a partial agonist drug for μ-type opioid peptide receptors (MORs), and by combining quantitative mass spectrometry with cell biological analysis to investigate the reduced efficacy of morphine, compared to that of a peptide full agonist, in promoting receptor endocytosis. We showed that these chemically distinct ligands produced a complex and qualitatively similar mixture of phosphorylated opioid receptor forms in intact cells. Quantitatively, however, the different agonists promoted disproportionate multisite phosphorylation of a specific serine and threonine motif, and we found that modification at more than one residue was essential for the efficient recruitment of the adaptor protein β-arrestin that mediated subsequent endocytosis of MORs. Thus, quantitative encoding of agonist-selective endocytosis at the level of individual opioid receptors was based on the conserved biochemical principles of multisite phosphorylation and threshold detection.
Collapse
Affiliation(s)
- Elaine K Lau
- Department of Psychiatry, University of California, San Francisco, CA 94158, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Raehal KM, Schmid CL, Groer CE, Bohn LM. Functional selectivity at the μ-opioid receptor: implications for understanding opioid analgesia and tolerance. Pharmacol Rev 2011; 63:1001-19. [PMID: 21873412 PMCID: PMC3186080 DOI: 10.1124/pr.111.004598] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids are the most effective analgesic drugs for the management of moderate or severe pain, yet their clinical use is often limited because of the onset of adverse side effects. Drugs in this class produce most of their physiological effects through activation of the μ opioid receptor; however, an increasing number of studies demonstrate that different opioids, while presumably acting at this single receptor, can activate distinct downstream responses, a phenomenon termed functional selectivity. Functional selectivity of receptor-mediated events can manifest as a function of the drug used, the cellular or neuronal environment examined, or the signaling or behavioral measure recorded. This review summarizes both in vitro and in vivo work demonstrating functional selectivity at the μ opioid receptor in terms of G protein coupling, receptor phosphorylation, interactions with β-arrestins, receptor desensitization, internalization and signaling, and details on how these differences may relate to the progression of analgesic tolerance after their extended use.
Collapse
Affiliation(s)
- Kirsten M Raehal
- Molecular Therapeutics and Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | | | | | | |
Collapse
|
39
|
Groer CE, Schmid CL, Jaeger AM, Bohn LM. Agonist-directed interactions with specific beta-arrestins determine mu-opioid receptor trafficking, ubiquitination, and dephosphorylation. J Biol Chem 2011; 286:31731-41. [PMID: 21757712 DOI: 10.1074/jbc.m111.248310] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Morphine and other opiates mediate their effects through activation of the μ-opioid receptor (MOR), and regulation of the MOR has been shown to critically affect receptor responsiveness. Activation of the MOR results in receptor phosphorylation, β-arrestin recruitment, and internalization. This classical regulatory process can differ, depending on the ligand occupying the receptor. There are two forms of β-arrestin, β-arrestin1 and β-arrestin2 (also known as arrestin2 and arrestin3, respectively); however, most studies have focused on the consequences of recruiting β-arrestin2 specifically. In this study, we examine the different contributions of β-arrestin1- and β-arrestin2-mediated regulation of the MOR by comparing MOR agonists in cells that lack expression of individual or both β-arrestins. Here we show that morphine only recruits β-arrestin2, whereas the MOR-selective enkephalin [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]enkephalin (DAMGO), recruits either β-arrestin. We show that β-arrestins are required for receptor internalization and that only β-arrestin2 can rescue morphine-induced MOR internalization, whereas either β-arrestin can rescue DAMGO-induced MOR internalization. DAMGO activation of the receptor promotes MOR ubiquitination over time. Interestingly, β-arrestin1 proves to be critical for MOR ubiquitination as modification does not occur in the absence of β-arrestin1 nor when morphine occupies the receptor. Moreover, the selective interactions between the MOR and β-arrestin1 facilitate receptor dephosphorylation, which may play a role in the resensitization of the MOR and thereby contribute to overall development of opioid tolerance.
Collapse
Affiliation(s)
- Chad E Groer
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | | | | | | |
Collapse
|
40
|
Patierno S, Anselmi L, Jaramillo I, Scott D, Garcia R, Sternini C. Morphine induces μ opioid receptor endocytosis in guinea pig enteric neurons following prolonged receptor activation. Gastroenterology 2011; 140:618-26. [PMID: 21070774 PMCID: PMC3033567 DOI: 10.1053/j.gastro.2010.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2010] [Revised: 10/18/2010] [Accepted: 11/02/2010] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS The μ opioid receptor (μOR) undergoes rapid endocytosis after acute stimulation with opioids and most opiates, but not with morphine. We investigated whether prolonged activation of μOR affects morphine's ability to induce receptor endocytosis in enteric neurons. METHODS We compared the effects of morphine, a poor μOR-internalizing opiate, and (D-Ala2,MePhe4,Gly-ol5) enkephalin (DAMGO), a potent μOR-internalizing agonist, on μOR trafficking in enteric neurons and on the expression of dynamin and β-arrestin immunoreactivity in the ileum of guinea pigs rendered tolerant by chronic administration of morphine. RESULTS Morphine (100 μmol/L) strongly induced endocytosis of μOR in tolerant but not naive neurons (55.7% ± 9.3% vs 24.2% ± 7.3%; P < .001) whereas DAMGO (10 μmol/L) strongly induced internalization of μOR in neurons from tolerant and naive animals (63.6% ± 8.4% and 66.5% ± 3.6%). Morphine- or DAMGO-induced μOR endocytosis resulted from direct interactions between the ligand and the μOR because endocytosis was not affected by tetrodotoxin, a blocker of endogenous neurotransmitter release. Ligand-induced μOR internalization was inhibited by pretreatment with the dynamin inhibitor, dynasore. Chronic morphine administration resulted in a significant increase and translocation of dynamin immunoreactivity from the intracellular pool to the plasma membrane, but did not affect β-arrestin immunoreactivity. CONCLUSIONS Chronic activation of μORs increases the ability of morphine to induce μOR endocytosis in enteric neurons, which depends on the level and cellular localization of dynamin, a regulatory protein that has an important role in receptor-mediated signal transduction in cells.
Collapse
Affiliation(s)
- Simona Patierno
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Laura Anselmi
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Ingrid Jaramillo
- Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
| | - David Scott
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Physiology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Rachel Garcia
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Catia Sternini
- CURE Digestive Diseases Research Center, Veterans Administration Greater Los Angeles Healthcare System, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, Digestive Diseases Division, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
41
|
von Zastrow M. Regulation of opioid receptors by endocytic membrane traffic: mechanisms and translational implications. Drug Alcohol Depend 2010; 108:166-71. [PMID: 20338697 PMCID: PMC3417350 DOI: 10.1016/j.drugalcdep.2010.02.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 02/17/2010] [Accepted: 02/19/2010] [Indexed: 11/27/2022]
Abstract
Opioid neuropeptide receptors mediate diverse physiological functions and are important targets for both therapeutic and abused drugs. Opioid receptors are highly regulated in intact cells, and there is reason to believe that this regulation controls the clinical effects of opioid drugs. The present review will discuss some of this evidence, focusing specifically on the regulation of opioid receptors by endocytic membrane trafficking mechanisms. First, some basic principles of regulated endocytosis will be reviewed, and the principle of 'molecular sorting' as a means to determine the functional consequences of endocytosis will be introduced, Most of this information has been derived from studies of simplified cell models. Second, present knowledge about the operation of these mechanisms in physiologically relevant CNS neurons will be discussed, focusing on studies of neurons cultured from rodent brain. Third, recent insight into the effects of endocytic trafficking on opioid regulation in vivo will be considered, focusing on results from studies of transgenic mouse models. Much remains to be learned at these pre-clinical levels, and effects of endocytosis on opioid actions in humans remain completely unexplored. Two particular insights, which have emerged from pre-clinical studies, will be proposed for translational consideration.
Collapse
Affiliation(s)
- Mark von Zastrow
- Department of Psychiatry, N212E Genentech Hall, UCSF Mission Bay Campus, San Francisco, CA 94158-2140, USA.
| |
Collapse
|
42
|
Walwyn WM, Miotto KA, Evans CJ. Opioid pharmaceuticals and addiction: the issues, and research directions seeking solutions. Drug Alcohol Depend 2010; 108:156-65. [PMID: 20188495 PMCID: PMC3072810 DOI: 10.1016/j.drugalcdep.2010.01.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 12/29/2009] [Accepted: 01/08/2010] [Indexed: 11/21/2022]
Abstract
There are few pharmaceuticals superior to opiates for the treatment of pain. However, with concerns of addiction, withdrawal and questionable efficacy for all types of pain, these compounds are far from a magical panacea for pain-relief. As it is unlikely that other classes of compounds will supersede the opioids in the very near future, it is important to both optimize current opioid therapies and curb the astounding diversion of opioids from their intended analgesic use to non-medical abuse. In optimizing opioid therapeutics it is necessary to enhance the clinical awareness of the benefits of treating pain and combine this with aggressive strategies to reduce diversion for non-medical use. At the heart of the issue of opioid misuse is the role of opioid systems in the reward circuitry, and the adaptive processes associated with repetitive opioid use that manifest during withdrawal. Emerging pharmacological insights of opioid receptors will be reviewed that provide future hope for developing opioid-based analgesics with reduced addictive properties and perhaps, reduced opponent processes. In addition, with the increased understanding of nociceptive circuitry and the molecules involved in transmitting pain, new therapeutic targets have become evident that may result in effective analgesics either alone or in combination with current opioid therapies.
Collapse
Affiliation(s)
- Wendy M Walwyn
- Shirley and Stephan Hatos Center for Neuropharmacology, Semel Institute, Los Angeles, CA 90024-1759, USA.
| | | | | |
Collapse
|
43
|
Margas W, Mahmoud S, Ruiz-Velasco V. Muscarinic acetylcholine receptor modulation of mu (mu) opioid receptors in adult rat sphenopalatine ganglion neurons. J Neurophysiol 2009; 103:172-82. [PMID: 19889856 DOI: 10.1152/jn.00295.2009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The sphenopalatine ganglion (SPG) neurons represent the parasympathetic branch of the autonomic nervous system involved in controlling cerebral blood flow. In the present study, we examined the coupling mechanism between mu (mu) opioid receptors (MOR) and muscarinic acetylcholine receptors (mAChR) with Ca(2+) channels in acutely dissociated adult rat SPG neurons. Successful MOR activation was recorded in approximately 40-45% of SPG neurons employing the whole cell variant of the patch-clamp technique. In addition, immunofluorescence assays indicated that MOR are not expressed in all SPG neurons while M(2) mAChR staining was evident in all neurons. The concentration-response relationships generated with morphine and [d-Ala2-N-Me-Phe4-Glycol5]-enkephalin (DAMGO) showed IC(50) values of 15.2 and 56.1 nM and maximal Ca(2+) current inhibition of 26.0 and 38.7%, respectively. Activation of MOR or M(2) mAChR with morphine or oxotremorine-methiodide (Oxo-M), respectively, resulted in voltage-dependent inhibition of Ca(2+) currents via coupling with Galpha(i/o) protein subunits. The acute prolonged exposure (10 min) of neurons to morphine or Oxo-M led to the homologous desensitization of MOR and M(2) mAChR, respectively. The prolonged stimulation of M(2) mAChR with Oxo-M resulted in heterologous desensitization of morphine-mediated Ca(2+) current inhibition, and was sensitive to the M(2) mAChR blocker methoctramine. On the other hand, when the neurons were exposed to morphine or DAMGO for 10 min, heterologous desensitization of M(2) mAChR was not observed. These results suggest that in rat SPG neurons activation of M(2) mAChR likely modulates opioid transmission in the brain vasculature to adequately maintain cerebral blood flow.
Collapse
Affiliation(s)
- Wojciech Margas
- Department of Anesthesiology, Penn State University College of Medicine, 500 University Drive, Hershey, PA 17033-0850, USA
| | | | | |
Collapse
|
44
|
Lin H, Higgins P, Loh HH, Law PY, Liao D. Bidirectional effects of fentanyl on dendritic spines and AMPA receptors depend upon the internalization of mu opioid receptors. Neuropsychopharmacology 2009; 34:2097-111. [PMID: 19295508 PMCID: PMC2731771 DOI: 10.1038/npp.2009.34] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fentanyl is a frequently used and abused opioid analgesic and can cause internalization of mu opioid receptors (MORs). Receptor internalization modulates the signaling pathways of opioid receptors. As changes in dendritic spines and synaptic AMPA receptors play important roles in addiction and memory loss, we investigated how fentanyl affects dendritic spines and synaptic AMPA receptors in cultured hippocampal neurons. Fentanyl at low concentrations (0.01 and 0.1 microM) caused the collapse of dendritic spines and decreased the number of AMPA receptor clusters. In contrast, fentanyl at high concentrations (1 and 10 microM) had opposite effects, inducing the emergence of new spines and increasing the number of AMPA receptor clusters. These dose-dependent bidirectional effects of fentanyl were blocked by a selective MOR antagonist CTOP at 5 microM. In neurons that had been transfected with HA-tagged or GFP-tagged MORs, fentanyl at high concentrations induced persistent and robust internalization of MORs, whereas fentanyl at lower concentrations induced little or transient receptor internalization. The blockade of receptor internalization with the expression of dominant-negative Dynamin I (the K44E mutant) reversed the effect of fentanyl at high concentrations, supporting a role of receptor internalization in modulating the dose-dependent effects of fentanyl. In contrast to morphine, the effects of fentanyl on dendritic spines are distinctively bidirectional and concentration dependent, probably due to its ability to induce robust internalization of MORs at high concentrations. The characterization of the effects of fentanyl on spines and AMPA receptors may help us understand the roles of MOR internalization in addiction and cognitive deficits.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Animals
- Cell Membrane/drug effects
- Cell Membrane/physiology
- Cells, Cultured
- Central Nervous System Agents
- Dendritic Spines/drug effects
- Dendritic Spines/physiology
- Dose-Response Relationship, Drug
- Dynamin I/genetics
- Dynamin I/metabolism
- Fentanyl/administration & dosage
- Fentanyl/pharmacology
- Hippocampus/drug effects
- Hippocampus/physiology
- Mutation, Missense
- Neurons/drug effects
- Neurons/physiology
- Protein Transport/drug effects
- Rats
- Receptors, AMPA/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Somatostatin/analogs & derivatives
- Somatostatin/pharmacology
- Synapses/drug effects
- Synapses/physiology
Collapse
Affiliation(s)
- Hang Lin
- Department of Neuroscience, The University of Minnesota, 321 Church St S.E. Minneapolis, MN 55455
- Department of Neurology, Chengdu General Military Hospital, Chengdu City, 610083, China
| | - Paul Higgins
- Department of Neuroscience, The University of Minnesota, 321 Church St S.E. Minneapolis, MN 55455
| | - Horace H. Loh
- Department of Pharmacology, The University of Minnesota, 321 Church St S.E. Minneapolis, MN 55455
| | - Ping-Yee Law
- Department of Pharmacology, The University of Minnesota, 321 Church St S.E. Minneapolis, MN 55455
| | - Dezhi Liao
- Department of Neuroscience, The University of Minnesota, 321 Church St S.E. Minneapolis, MN 55455
| |
Collapse
|
45
|
Bodzon-Kulakowska A, Suder P, Mak P, Bierczynska-Krzysik A, Lubec G, Walczak B, Kotlinska J, Silberring J. Proteomic analysis of striatal neuronal cell cultures after morphine administration. J Sep Sci 2009; 32:1200-10. [PMID: 19296477 DOI: 10.1002/jssc.200800464] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Using primary neuronal cell culture assays, combined with 2-D gel electrophoresis and capillary LC-MS, we identified differences in proteomes between control and morphine-treated cells. Statistically significant differences were observed among 26 proteins. Nineteen of them were up-regulated, while seven were down-regulated in morphine-treated cell populations. The identified proteins belong to classes involved in energy metabolism, associated with oxidative stress, linked with protein biosynthesis, cytoskeletal ones, and chaperones. The detected proteins demand further detailed studies of their biological roles in morphine addiction. It is crucial to confirm observed processes in vivo in order to reveal the nature and importance of the biological effect of proteome changes after morphine administration. Further investigations may lead to the discovery of new proteome-based effects of morphine on living organisms.
Collapse
Affiliation(s)
- Anna Bodzon-Kulakowska
- Neurobiochemistry Department, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Functional characterization of human variants of the mu-opioid receptor gene. Proc Natl Acad Sci U S A 2009; 106:10811-6. [PMID: 19528663 DOI: 10.1073/pnas.0904509106] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Opioids and their receptors have an important role in analgesia and alcohol and substance use disorders (ASUD). We have identified several naturally occurring amino acid changing variants of the human mu-opioid receptor (MOR), and assessed the functional consequences of these previously undescribed variants in stably expressing cell lines. Several of these variants had altered trafficking and signaling properties. We found that an L85I variant showed significant internalization in response to morphine, in contrast to the WT MOR, which did not internalize in response to morphine. Also, when L85I and WT receptor were coexpressed, WT MOR internalized with the L85I MOR, suggesting that, in the heterozygous condition, the L85I phenotype would be dominant. This finding is potentially important, because receptor internalization has been associated with development of tolerance to opiate analgesics. In contrast, an R181C variant abolished both signaling and internalization in response to saturating doses of the hydrolysis-resistant enkephalin [D-Ala2,N-MePhe4,Gly5-ol]enkephalin (DAMGO). Coexpression of the R181C and WT receptor led to independent trafficking of the 2 receptors. S42T and C192F variants showed a rightward shift in potency of both morphine and DAMGO, whereas the S147C variant displayed a subtle leftward shift in morphine potency. These data suggest that these and other such variants may have clinical relevance to opioid responsiveness to both endogenous ligands and exogenous drugs, and could influence a broad range of phenotypes, including ASUD, pain responses, and the development of tolerance to morphine.
Collapse
|
47
|
Zhang Y, Xiong W, Lin X, Ma X, Yu LC. Receptor trafficking induced by mu-opioid-receptor phosphorylation. Neurosci Biobehav Rev 2009; 33:1192-7. [PMID: 19747597 DOI: 10.1016/j.neubiorev.2009.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 03/10/2009] [Accepted: 03/11/2009] [Indexed: 10/21/2022]
Abstract
Opiates, including morphine, are widely used drugs for antinociception in clinics. Prolonged treatments of opioids induce both tolerance and dependence, which are the major side effects of opioid therapy. One of the mechanisms for the development of tolerance and dependence is implicated to be opioid-receptor trafficking. Here we review the current understandings of opioid-receptor phosphorylation, endocytosis and desensitization after repeated agonist treatments. Also, the role of G-protein coupled receptor kinases in opioid-receptor phosphorylation is discussed. How to associate these observations to physiological and behavioral changes in animal models and clinics is still under investigation.
Collapse
Affiliation(s)
- Yan Zhang
- Laboratory of Neurobiology, College of Life Sciences, Peking University, Beijing 100871, China.
| | | | | | | | | |
Collapse
|
48
|
Scavone JL, Van Bockstaele EJ. Mu-opioid receptor redistribution in the locus coeruleus upon precipitation of withdrawal in opiate-dependent rats. Anat Rec (Hoboken) 2009; 292:401-11. [PMID: 19248160 PMCID: PMC2863286 DOI: 10.1002/ar.20860] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Administration of mu-opioid receptor (MOR) agonists is known to produce adaptive changes within noradrenergic neurons of the rat locus coeruleus (LC). Alterations in the subcellular distribution of MOR have been shown to occur in the LC in response to full agonists and endogenous peptides; however, there is considerable debate in the literature whether trafficking of MOR occurs after chronic exposure to the partial-agonist morphine. In the present study, we examined adaptations in MOR after chronic opioid exposure using immunofluorescence and electron microscopy (EM), using receptor internalization as a functional endpoint. MOR trafficking in LC neurons was characterized in morphine-dependent rats that were given naltrexone at a dose known to precipitate withdrawal. After chronic morphine exposure, a subtle redistribution of MOR immunoreactivity from the membrane to the cytosol was detected within dendrites of LC neurons. Interestingly, an acute injection of naltrexone in rats exposed to chronic morphine produced a robust internalization of MOR, whereas administration of naltrexone failed to do so in naïve animals. These findings provide anatomical evidence for modified regulation of MOR trafficking after chronic morphine treatment in brain noradrenergic neurons. Adaptations in the MOR signaling pathways that regulate internalization may occur as a consequence of chronic treatment and precipitation of withdrawal. Mechanisms underlying this effect might include differential MOR regulation in the LC, or downstream effects of withdrawal-induced enkephalin (ENK) release from afferents to the LC.
Collapse
Affiliation(s)
- Jillian L Scavone
- Department of Neurosurgery, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylania, USA
| | | |
Collapse
|
49
|
Wang Y, Van Bockstaele EJ, Liu-Chen LY. In vivo trafficking of endogenous opioid receptors. Life Sci 2008; 83:693-9. [PMID: 18930741 PMCID: PMC2652254 DOI: 10.1016/j.lfs.2008.09.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Revised: 09/11/2008] [Accepted: 09/17/2008] [Indexed: 12/14/2022]
Abstract
Several approaches have been taken for these in vivo studies. In many studies, the use of semi-quantitative immuno-electron microscopy is the approach of choice. Endogenous opioid receptors display differential subcellular distributions with mu opioid receptor (MOPR) being mostly present on the plasma membrane and delta-opioid receptor (DOPR) and kappa-opioid receptor (KOPR) having a significant intracellular pool. Etorphine and DAMGO cause endocytosis of the MOPR, but morphine does not, except in some dendrites. Interestingly, chronic inflammatory pain and morphine treatment promote trafficking of intracellular DOPR to the cell surface which may account for the enhanced antinociceptive effects of DOPR agonists. KOPR has been reported to be associated with secretory vesicles in the posterior pituitary and translocated to the cell surface upon salt loading along with the release of vasopressin. The study of endogenous opioid receptors using in vivo models has produced some interesting results that could not have been anticipated in vitro. In vivo studies, therefore, are essential to provide insight into the mechanisms underlying opioid receptor regulation.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Humans
- Opioid Peptides/drug effects
- Opioid Peptides/genetics
- Opioid Peptides/physiology
- Receptors, Cell Surface/drug effects
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/physiology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Signal Transduction
- Subcellular Fractions
Collapse
Affiliation(s)
- Yulin Wang
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| | - Elisabeth J. Van Bockstaele
- Department of Neurosurgery, Farber Institute for Neurosciences, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - Lee-Yuan Liu-Chen
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
50
|
Tanowitz M, Hislop JN, von Zastrow M. Alternative splicing determines the post-endocytic sorting fate of G-protein-coupled receptors. J Biol Chem 2008; 283:35614-21. [PMID: 18936093 DOI: 10.1074/jbc.m806588200] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mu-type opioid receptors are physiologically important G-protein-coupled receptors that are generally thought to recycle after agonist-induced endocytosis. Here we show that several alternatively spliced receptor variants fail to do so efficiently because of splice-mediated removal of an endocytic sorting sequence that is present specifically in the MOR1 variant. All of the recycling-impaired receptor variants were found to undergo proteolytic down-regulation more rapidly than MOR1, irrespective of moderate differences in endocytic rate, indicating that alternative splicing plays a specific role in distinguishing the trafficking itinerary of receptors after endocytosis. The recycling-impaired MOR1B variant was similar to MOR1 in its ability to mediate opioid-dependent inhibition of adenylyl cyclase, and to undergo opioid-induced desensitization in intact cells. Functional recovery (resensitization) of MOR1B-mediated cellular responsiveness after opioid removal, however, was significantly impaired (4-fold reduction in rate) compared with MOR1. To our knowledge the present results are the first to establish a role of alternative RNA processing in specifying the post-endocytic sorting of G-protein-coupled receptors between divergent and functionally distinct membrane pathways.
Collapse
Affiliation(s)
- Michael Tanowitz
- Department of Psychiatry, University of California, San Francisco, California 94158-2140, USA
| | | | | |
Collapse
|