1
|
Xu Y, Shao N, Zhi F, Chen R, Yang Y, Li J, Xia Y, Peng Y. Delta-opioid receptor signaling alleviates neuropathology and cognitive impairment in the mouse model of Alzheimer's disease by regulating microglia homeostasis and inhibiting HMGB1 pathway. Alzheimers Res Ther 2025; 17:35. [PMID: 39893485 PMCID: PMC11786345 DOI: 10.1186/s13195-025-01682-1] [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: 11/20/2024] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Recent studies suggest that opioid receptor signaling may differentially affect Alzheimer's disease (AD) pathology and the relevant behavioral dysfunctions. However, the precise roles and mechanisms of opioid receptor subtypes in AD pathologies are still unclear with major controversies. METHODS We compared the delta-opioid receptor (DOR)- and mu-opioid receptor (MOR)-mediated effects on AD-associated cognitive deficits, pathologies, neuroinflammations, cell death using transgenic APP/PS1 mouse model and BV2 cell line at behavioral, molecular, and cellular levels. Unpaired t-test and one/two way analysis for variance (ANOVA) were used to analyze statistical significance of the data. RESULTS We show a distinct role of DOR and its major difference with MOR in AD injury in an APP/PS1 mouse model. DOR activation by UFP-512, but not MOR activation by DAMGO, attenuated cognitive impairment, reduced beta-amyloid (Aβ) production and aggregation, as well as protected the neurons from apoptosis in APP/PS1 mice. DOR and MOR also differentially modulated microglia in APP/PS1 mice and in vitro AD cell model with a DOR-mediated inhibition on the excessive activation of microglia and the release of pro-inflammatory cytokines in AD pathologies. Gene expression profiling further revealed that the alternations in DOR/MOR are closely associated with microglial homeostatic signatures and high mobility group protein B1 (HMGB1) in AD. DOR activation inhibited HMGB1 secretion and its translocation from nuclear to cytoplasm. Our in-vitro studies further confirmed that DOR overexpression mitigated microglial inflammatory response and rescued neurons from AD injury via HMGB1-NF-κB signaling pathway. CONCLUSIONS These novel findings uncover previously unappreciated roles of DOR in neuroprotection against AD injury via modulating microglia-related inflammatory responses.
Collapse
Affiliation(s)
- Yuan Xu
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Naiyuan Shao
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Feng Zhi
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ronghua Chen
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Yilin Yang
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jiahui Li
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
- Shanghai Research Center for Acupuncture and Meridians, Shanghai, China
| | - Ying Xia
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China.
- Shanghai Research Center for Acupuncture and Meridians, Shanghai, China.
| | - Ya Peng
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China.
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China.
| |
Collapse
|
2
|
Namchuk AB, Tsuda MC, Lucki I, Browne CA. Kappa opioid receptor mediated operant performance in male and female rats. Pharmacol Biochem Behav 2024; 244:173847. [PMID: 39151827 DOI: 10.1016/j.pbb.2024.173847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Anhedonia and avolition are emotions frequently endorsed by individuals with stress related disorders. Kappa opioid receptor (KOR) activation can induce negative emotions and recent clinical evidence suggests that KOR antagonism can alleviate anhedonia in a transdiagnostic cohort of patients. However, the behavioral consequences of KOR activation and antagonism in modulating motivation, as assessed by schedule-controlled behavioral performance without preexisting conditions (stress or substance use), have not been formally assessed. To address this gap in the literature, this report utilized male and female Sprague Dawley rats to (1) evaluate the impact of the selective KOR agonist U50,488, on the performance of animals responding for sucrose pellets under a progressive ratio (PR) schedule and (2) determine the effects of the short-acting KOR antagonist LY2444296 alone and on U50,488 mediated reductions in PR performance. Overall, U50,488 5 mg/kg significantly reduced the breakpoint and number of rewards obtained by animals. This occurred in the absence of motor impairment and independent of evidence for satiation. LY2444296 did not alter PR performance when administered alone but effectively blocked the deficits induced by U50,488. To further delineate the behavioral alterations that underlie these reductions in responding, a more detailed analysis was conducted on PR performance in the first 15 min of the session, the period of time when animals obtained the most reinforcers. During this period, U50,488 increased the length of the post-reinforcement pause and reduced the running rate on PR schedules. These changes in behavior produced by acute activation of KORs are consistent with a reduction of effort-related motivation in rodents. These data contribute to the understanding of how KORs modulate motivation, which is critical to future efforts to evaluate performance in the context of stress and assess how KOR antagonists alleviate anhedonic behaviors associated with stress.
Collapse
Affiliation(s)
- Amanda B Namchuk
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Mumeko C Tsuda
- Preclinical Behavior & Modelling Core, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America; Preclinical Behavior & Modelling Core, Uniformed Services University, Bethesda, MD 20814, United States of America; Department of Psychiatry, Uniformed Services University, Bethesda, MD 20814, United States of America
| | - Caroline A Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD 20814, United States of America.
| |
Collapse
|
3
|
Goode-Romero G, Dominguez L. Descriptive molecular pharmacology of the δ opioid receptor (DOR): A computational study with structural approach. PLoS One 2024; 19:e0304068. [PMID: 38991032 PMCID: PMC11239112 DOI: 10.1371/journal.pone.0304068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 05/06/2024] [Indexed: 07/13/2024] Open
Abstract
This work focuses on the δ receptor (DOR), a G protein-coupled receptor (GPCR) belonging to the opioid receptor group. DOR is expressed in numerous tissues, particularly within the nervous system. Our study explores computationally the receptor's interactions with various ligands, including opiates and opioid peptides. It elucidates how these interactions influence the δ receptor response, relevant in a wide range of health and pathological processes. Thus, our investigation aims to explore the significance of DOR as an incoming drug target for pain relief and neurodegenerative diseases and as a source for novel opioid non-narcotic analgesic alternatives. We analyze the receptor's structural properties and interactions using Molecular Dynamics (MD) simulations and Gaussian-accelerated MD across different functional states. To thoroughly assess the primary differences in the structural and conformational ensembles across our different simulated systems, we initiated our study with 1 μs of conventional Molecular Dynamics. The strategy was chosen to encompass the full activation cycle of GPCRs, as activation processes typically occur within this microsecond range. Following the cMD, we extended our study with an additional 100 ns of Gaussian accelerated Molecular Dynamics (GaMD) to enhance the sampling of conformational states. This simulation approach allowed us to capture a comprehensive range of dynamic interactions and conformational changes that are crucial for GPCR activation as influenced by different ligands. Our study includes comparing agonist and antagonist complexes to uncover the collective patterns of their functional states, regarding activation, blocking, and inactivation of DOR, starting from experimental data. In addition, we also explored interactions between agonist and antagonist molecules from opiate and opioid classifications to establish robust structure-activity relationships. These interactions have been systematically quantified using a Quantitative Structure-Activity Relationships (QSAR) model. This research significantly contributes to our understanding of this significant pharmacological target, which is emerging as an attractive subject for drug development.
Collapse
Affiliation(s)
- Guillermo Goode-Romero
- Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Laura Dominguez
- Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
4
|
Hebert SV, Green MA, Mashaw SA, Brouillette WD, Nguyen A, Dufrene K, Shelvan A, Patil S, Ahmadzadeh S, Shekoohi S, Kaye AD. Assessing Risk Factors and Comorbidities in the Treatment of Chronic Pain: A Narrative Review. Curr Pain Headache Rep 2024; 28:525-534. [PMID: 38558165 DOI: 10.1007/s11916-024-01249-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW Chronic pain affects a significant portion of the population globally, making it a leading cause of disability. Understanding the multifaceted nature of chronic pain, its various types, and the intricate relationship it shares with risk factors, comorbidities, and mental health issues like depression and anxiety is critical for comprehensive patient care. Factors such as socioeconomic status (SES), age, gender, and obesity collectively add layers of complexity to chronic pain experiences and pose management challenges. RECENT FINDINGS Low SES presents barriers to effective pain care, while gender differences and the prevalence of chronic pain in aging adults emphasize the need for tailored approaches. The association between chronic pain and physical comorbidities like cardiovascular disease, chronic obstructive pulmonary disease (COPD), and diabetes mellitus reveals shared risk factors and further highlights the importance of integrated treatment strategies. Chronic pain and mental health are intricately linked through biochemical mechanisms, profoundly affecting overall quality of life. This review explores pharmacologic treatment for chronic pain, particularly opioid analgesia, with attention to the risk of substance misuse and the ongoing opioid epidemic. We discuss the potential role of medical cannabis as an alternative treatment with a nuanced perspective on its impact on opioid use. Addressing the totality and complexity of pain states is crucial to individualizing chronic pain management. With different types of pain having different underlying mechanisms, considerations should be made when approaching their treatment. Moreover, the synergistic relationship that pain states can have with other comorbidities further complicates chronic pain conditions.
Collapse
Affiliation(s)
- Sage V Hebert
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Melanie A Green
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Sydney A Mashaw
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - William D Brouillette
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Angela Nguyen
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Kylie Dufrene
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Anitha Shelvan
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Shilpadevi Patil
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA.
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, 71103, USA
- Department of Pharmacology, Louisiana State University Health Sciences Center at Shreveport, Toxicology, and Neurosciences, Shreveport, LA, 71103, USA
| |
Collapse
|
5
|
Aslam MM, Fan KH, Lawrence E, Bedison MA, Snitz BE, DeKosky ST, Lopez OL, Feingold E, Kamboh MI. Genome-wide analysis identifies novel loci influencing plasma apolipoprotein E concentration and Alzheimer's disease risk. Mol Psychiatry 2023; 28:4451-4462. [PMID: 37666928 PMCID: PMC10827653 DOI: 10.1038/s41380-023-02170-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/16/2023] [Accepted: 06/27/2023] [Indexed: 09/06/2023]
Abstract
The APOE 2/3/4 polymorphism is the greatest genetic risk factor for Alzheimer's disease (AD). This polymorphism is also associated with variation in plasma ApoE level; while APOE*4 lowers, APOE*2 increases ApoE level. Lower plasma ApoE level has also been suggested to be a risk factor for incident dementia. To our knowledge, no large genome-wide association study (GWAS) has been reported on plasma ApoE level. This study aimed to identify new genetic variants affecting plasma ApoE level as well as to test if baseline ApoE level is associated with cognitive function and incident dementia in a longitudinally followed cohort of the Ginkgo Evaluation of Memory (GEM) study. Baseline plasma ApoE concentration was measured in 3031 participants (95.4% European Americans (EAs)). GWAS analysis was performed on 2580 self-identified EAs where both genotype and plasma ApoE data were available. Lower ApoE concentration was associated with worse cognitive function, but not with incident dementia. As expected, the risk for AD increased from E2/2 through to E4/4 genotypes (P for trend = 4.8E-75). In addition to confirming the expected and opposite associations of APOE*2 (P = 4.73E-79) and APOE*4 (P = 8.73E-12) with ApoE level, GWAS analysis revealed nine additional independent signals in the APOE region, and together they explained about 22% of the variance in plasma ApoE level. We also identified seven new loci on chromosomes 1, 4, 5, 7, 11, 12 and 20 (P range = 5.49E-08 to 5.36E-10) that explained about 9% of the variance in ApoE level. Plasma ApoE level-associated independent variants, especially in the APOE region, were also associated with AD risk and amyloid deposition in the brain, indicating that genetically determined ApoE level variation may be a risk factor for developing AD. These results improve our understanding of the genetic determinants of plasma ApoE level and their potential value in affecting AD risk.
Collapse
Affiliation(s)
- M Muaaz Aslam
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kang-Hsien Fan
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Elizabeth Lawrence
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Margaret Anne Bedison
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beth E Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven T DeKosky
- McKnight Brain Institute and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Oscar L Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eleanor Feingold
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Tanguturi P, Streicher JM. The role of opioid receptors in modulating Alzheimer's Disease. Front Pharmacol 2023; 14:1056402. [PMID: 36937877 PMCID: PMC10014470 DOI: 10.3389/fphar.2023.1056402] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Alzheimer's disease (AD) is a complex neurological disorder characterized by accumulation of amyloid plaques and neurofibrillary tangles. Long term investigation of AD pathogenesis suggests that β-site amyloid precursor protein [APP] cleaving enzyme 1 (BACE1) and γ-secretase enzymes promote the amyloidogenic pathway and produce toxic Aβ peptides that are predisposed to aggregate in the brain. Hence, the targeted inhibition of BACE1/γ-secretase expression and function is a promising approach for AD therapy. Several reports have suggested that the opioid family of G-protein coupled receptors modulate the etiology of AD progression. It has also been found that changes in the signaling pathways of opioid receptors increased the expression of BACE1 and γ-secretase, and is strongly correlated with abnormal production of Aβ and pathogenesis of AD. Thus, the opioid receptor family is a promising candidate for targeted drug development to treat AD. In this review, we outline the involvement and mechanisms of opioid receptor signaling modulation in Alzheimer's Disease progression.
Collapse
Affiliation(s)
- Parthasaradhireddy Tanguturi
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| | - John M. Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
7
|
Singh P, Singh D, Srivastava P, Mishra G, Tiwari AK. Evaluation of advanced, pathophysiologic new targets for imaging of CNS. Drug Dev Res 2023; 84:484-513. [PMID: 36779375 DOI: 10.1002/ddr.22040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/12/2022] [Accepted: 12/31/2022] [Indexed: 02/14/2023]
Abstract
The inadequate information about the in vivo pathological, physiological, and neurological impairments, as well as the absence of in vivo tools for assessing brain penetrance and the efficiency of newly designed drugs, has hampered the development of new techniques for the treatment for variety of new central nervous system (CNS) diseases. The searching sites such as Science Direct and PubMed were used to find out the numerous distinct tracers across 16 CNS targets including tau, synaptic vesicle glycoprotein, the adenosine 2A receptor, the phosphodiesterase enzyme PDE10A, and the purinoceptor, among others. Among the most encouraging are [18 F]FIMX for mGluR imaging, [11 C]Martinostat for Histone deacetylase, [18 F]MNI-444 for adenosine 2A imaging, [11 C]ER176 for translocator protein, and [18 F]MK-6240 for tau imaging. We also reviewed the findings for each tracer's features and potential for application in CNS pathophysiology and therapeutic evaluation investigations, including target specificity, binding efficacy, and pharmacokinetic factors. This review aims to present a current evaluation of modern positron emission tomography tracers for CNS targets, with a focus on recent advances for targets that have newly emerged for imaging in humans.
Collapse
Affiliation(s)
- Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Deepika Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Pooja Srivastava
- Division of Cyclotron and Radiopharmaceuticals Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Gauri Mishra
- Department of Zoology, Swami Shraddhananad College, University of Delhi, Alipur, Delhi, India
| | - Anjani K Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
8
|
Gallego-Villarejo L, Wallin C, Król S, Enrich-Bengoa J, Suades A, Aguilella-Arzo M, Gomara MJ, Haro I, Wärmlander S, Muñoz FJ, Gräslund A, Perálvarez-Marín A. Big dynorphin is a neuroprotector scaffold against amyloid β-peptide aggregation and cell toxicity. Comput Struct Biotechnol J 2022; 20:5672-5679. [PMID: 36284704 PMCID: PMC9582793 DOI: 10.1016/j.csbj.2022.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/09/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
Amyloid β-peptide (Aβ) misfolding into β-sheet structures triggers neurotoxicity inducing Alzheimer’s disease (AD). Molecules able to reduce or to impair Aβ aggregation are highly relevant as possible AD treatments since they should protect against Aβ neurotoxicity. We have studied the effects of the interaction of dynorphins, a family of opioid neuropeptides, with Aβ40 the most abundant species of Aβ. Biophysical measurements indicate that Aβ40 interacts with Big Dynorphin (BigDyn), lowering the amount of hydrophobic aggregates, and slowing down the aggregation kinetics. As expected, we found that BigDyn protects against Aβ40 aggregates when studied in human neuroblastoma cells by cell survival assays. The cross-interaction between BigDyn and Aβ40 provides insight into the mechanism of amyloid pathophysiology and may open up new therapy possibilities.
Collapse
Affiliation(s)
- Lucía Gallego-Villarejo
- Unit of Biophysics Dept of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Facultat de Medicina, 08193 Cerdanyola del Vallés, Catalonia, Spain
| | - Cecilia Wallin
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Sylwia Król
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Jennifer Enrich-Bengoa
- Unit of Biophysics Dept of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Facultat de Medicina, 08193 Cerdanyola del Vallés, Catalonia, Spain
| | - Albert Suades
- Unit of Biophysics Dept of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Facultat de Medicina, 08193 Cerdanyola del Vallés, Catalonia, Spain,Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Marcel Aguilella-Arzo
- Laboratory of Molecular Biophysics, Department of Physics, University Jaume I, 12071 Castellón, Spain
| | - María José Gomara
- Unitat de Síntesis i Aplicacions Biomèdiques de Pèptids, Institut de Química Avançada de Catalunya, IQAC-CSIC, Jordi Girona, 18-26, 08034 Barcelona, Catalonia, Spain
| | - Isabel Haro
- Unitat de Síntesis i Aplicacions Biomèdiques de Pèptids, Institut de Química Avançada de Catalunya, IQAC-CSIC, Jordi Girona, 18-26, 08034 Barcelona, Catalonia, Spain
| | - Sebastian Wärmlander
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Alex Perálvarez-Marín
- Unit of Biophysics Dept of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Facultat de Medicina, 08193 Cerdanyola del Vallés, Catalonia, Spain,Corresponding author.
| |
Collapse
|
9
|
Dalefield ML, Scouller B, Bibi R, Kivell BM. The Kappa Opioid Receptor: A Promising Therapeutic Target for Multiple Pathologies. Front Pharmacol 2022; 13:837671. [PMID: 35795569 PMCID: PMC9251383 DOI: 10.3389/fphar.2022.837671] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Kappa-opioid receptors (KOR) are widely expressed throughout the central nervous system, where they modulate a range of physiological processes depending on their location, including stress, mood, reward, pain, inflammation, and remyelination. However, clinical use of KOR agonists is limited by adverse effects such as dysphoria, aversion, and sedation. Within the drug-development field KOR agonists have been extensively investigated for the treatment of many centrally mediated nociceptive disorders including pruritis and pain. KOR agonists are potential alternatives to mu-opioid receptor (MOR) agonists for the treatment of pain due to their anti-nociceptive effects, lack of abuse potential, and reduced respiratory depressive effects, however, dysphoric side-effects have limited their widespread clinical use. Other diseases for which KOR agonists hold promising therapeutic potential include pruritis, multiple sclerosis, Alzheimer's disease, inflammatory diseases, gastrointestinal diseases, cancer, and ischemia. This review highlights recent drug-development efforts targeting KOR, including the development of G-protein-biased ligands, mixed opioid agonists, and peripherally restricted ligands to reduce side-effects. We also highlight the current KOR agonists that are in preclinical development or undergoing clinical trials.
Collapse
Affiliation(s)
| | | | | | - Bronwyn M. Kivell
- Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
10
|
Abstract
Temporal lobe epilepsy is considered to be one of the most common and severe forms of focal epilepsies. Patients frequently develop cognitive deficits and emotional blunting along progression of the disease. The high incidence of refractoriness to antiepileptic drugs and a frequent lack of admissibility to surgery pose an unmet medical challenge. In the urgent quest for novel treatment strategies, neuropeptides and their receptors are interesting candidates. However, their therapeutic potential has not yet been fully exploited. This chapter focuses on the functional role of the dynorphins (Dyns) and the kappa opioid receptor (KOR) system in temporal lobe epilepsy and the hippocampus.Genetic polymorphisms in the prepro-dynorphin (pDyn) gene causing lower levels of Dyns in humans and pDyn gene knockout in mice increase the risk to develop epilepsy. This suggests a role of Dyns and KOR as modulators of neuronal excitability. Indeed, KOR agonists induce inhibition of presynaptic neurotransmitter release, as well as postsynaptic hyperpolarization in glutamatergic neurons, both producing anticonvulsant effects.The development of new approaches to modulate the complex KOR signalling cascade (e.g. biased agonism and gene therapy) opens up new exciting therapeutic opportunities with regard to seizure control and epilepsy. Potential adverse side effects of KOR agonists may be minimized through functional selectivity or locally restricted treatment. Preclinical data suggest a high potential of such approaches to control seizures.
Collapse
Affiliation(s)
- Luca Zangrandi
- Institute of Virology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Freie Universität Berlin, Berlin, Germany
- Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
11
|
Lawn T, Aman Y, Rukavina K, Sideris-Lampretsas G, Howard M, Ballard C, Ray Chaudhuri K, Malcangio M. Pain in the neurodegenerating brain: insights into pharmacotherapy for Alzheimer disease and Parkinson disease. Pain 2021; 162:999-1006. [PMID: 33239526 PMCID: PMC7977618 DOI: 10.1097/j.pain.0000000000002111] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/18/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Timothy Lawn
- Centre for Neuroimaging Sciences, The Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Yahyah Aman
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Katarina Rukavina
- The Maurice Wohl Clinical Neuroscience Institute, The Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- Parkinson Foundation Centre of Excellence, King's College Hospital, London, United Kingdom
| | - George Sideris-Lampretsas
- Wolfson Centre for Age Related Diseases, The Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Matthew Howard
- Centre for Neuroimaging Sciences, The Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | | | - Kallol Ray Chaudhuri
- The Maurice Wohl Clinical Neuroscience Institute, The Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- Parkinson Foundation Centre of Excellence, King's College Hospital, London, United Kingdom
| | - Marzia Malcangio
- Wolfson Centre for Age Related Diseases, The Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
12
|
Babić Leko M, Hof PR, Šimić G. Alterations and interactions of subcortical modulatory systems in Alzheimer's disease. PROGRESS IN BRAIN RESEARCH 2021; 261:379-421. [PMID: 33785136 DOI: 10.1016/bs.pbr.2020.07.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) is not fully understood. Here we summarize current knowledge on the involvement of the serotonergic, noradrenergic, dopaminergic, cholinergic, and opioid systems in AD, emphasizing the importance of interactions between the serotonergic and the other subcortical modulatory systems during the progression of AD. In physiological conditions, all neurotransmitter systems function in concert and are interdependent at both the neuroanatomical and molecular levels. Through their early involvement in AD, cognitive and behavioral abilities that rely on their interactions also become disrupted. Considering that serotonin (5HT) regulates the release of noradrenaline (NA), dopamine (DA) and acetylcholine (ACh), any alteration in 5HT levels leads to disturbance of NA, DA, and ACh homeostasis in the brain. One of the earliest pathological changes during the prodromal phase of AD is a decrease of serotonergic transmission throughout the brain, with serotonergic receptors being also affected. Additionally, serotonergic and noradrenergic as well as serotonergic and dopaminergic nuclei are reciprocally interconnected. As the serotonergic dorsal raphe nucleus (DRN) is affected by pathological changes early in AD, and the noradrenergic locus coeruleus (LC) and dopaminergic ventral tegmental area (VTA) exhibit AD-related pathological changes, their connectivity also becomes altered in AD. Such disrupted interactions among neurotransmitter systems in AD can be used in the development of multi-target drugs. Some of the potential AD therapeutics (such as ASS234, RS67333, tropisetron) target multiple neurotransmitter systems to achieve the best possible improvement of cognitive and behavioral deficits observed in AD. Here, we review how serotonergic system interacts with other subcortical modulatory systems (noradrenergic, dopaminergic, cholinergic, and opioid systems) during AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia.
| |
Collapse
|
13
|
Pilozzi A, Carro C, Huang X. Roles of β-Endorphin in Stress, Behavior, Neuroinflammation, and Brain Energy Metabolism. Int J Mol Sci 2020; 22:E338. [PMID: 33396962 PMCID: PMC7796446 DOI: 10.3390/ijms22010338] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/23/2020] [Accepted: 12/26/2020] [Indexed: 12/25/2022] Open
Abstract
β-Endorphins are peptides that exert a wide variety of effects throughout the body. Produced through the cleavage pro-opiomelanocortin (POMC), β-endorphins are the primarily agonist of mu opioid receptors, which can be found throughout the body, brain, and cells of the immune system that regulate a diverse set of systems. As an agonist of the body's opioid receptors, β-endorphins are most noted for their potent analgesic effects, but they also have their involvement in reward-centric and homeostasis-restoring behaviors, among other effects. These effects have implicated the peptide in psychiatric and neurodegenerative disorders, making it a research target of interest. This review briefly summarizes the basics of endorphin function, goes over the behaviors and regulatory pathways it governs, and examines the variability of β-endorphin levels observed between normal and disease/disorder affected individuals.
Collapse
Affiliation(s)
| | | | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; (A.P.); (C.C.)
| |
Collapse
|
14
|
Xu Y, Zhi F, Balboni G, Yang Y, Xia Y. Opposite Roles of δ- and μ-Opioid Receptors in BACE1 Regulation and Alzheimer's Injury. Front Cell Neurosci 2020; 14:88. [PMID: 32425755 PMCID: PMC7204847 DOI: 10.3389/fncel.2020.00088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by amyloid plaques and neurofibrillary tangles. Substantial evidence for AD pathogenesis suggests that β-site APP cleaving enzyme 1 (BACE1) and γ-secretase enzyme initiate the amyloidogenic pathway and produces toxic Aβ peptides that prone to aggregate in the brain. Therefore, the inhibition of BACE1 expression and function is an attractive strategy for AD therapy. In the present work, we made the first finding that activating δ-opioid receptors (DOR) with a specific DOR agonist significantly attenuated BACE1 expression and activity in the highly differentiated PC12 cells with mimicked AD injury, while the application of DOR inhibitor naltrindole reversed the UFP-512 effects, and even caused a major increase in BACE1 expression and activity as well as Aβ42 production in physiological conditions. Knocking-down DOR also enhanced BACE1 protein expression and its activity for APP processing, associating with a significant increase in Aβ42 production. In sharp contrast, activation of μ-opioid receptor (MOR) with DAMGO greatly promoted BACE1 expression and activity with an acceleration of APP cleavage, thus contributing to increased Aβ42 production. DADLE, a less selective DOR agonist that may bind to MOR, had no stable inhibitory effect on BACE1. Similar results were also found in APP mutant (APPswe) SH-SY5Y cell line, providing further validation of the DOR action on BACE1 regulation. Our novel data demonstrated entirely different roles of DOR and MOR in the regulation of BACE1 expression and activity with DOR being neuroprotective against AD injury. These findings provided a novel clue for new strategies of AD therapy via targeting endogenous opioid receptors.
Collapse
Affiliation(s)
- Yuan Xu
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Department of Aeronautics and Astronautics, Fudan University, Shanghai, China.,Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, China
| | - Feng Zhi
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, China
| | - Gianfranco Balboni
- Department of Life and Environment Sciences, University of Cagliari, Cagliari, Italy
| | - Yilin Yang
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, China
| | - Ying Xia
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Department of Aeronautics and Astronautics, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Rocchi G, Sterlini B, Tardito S, Inglese M, Corradi A, Filaci G, Amore M, Magioncalda P, Martino M. Opioidergic System and Functional Architecture of Intrinsic Brain Activity: Implications for Psychiatric Disorders. Neuroscientist 2020; 26:343-358. [PMID: 32133917 DOI: 10.1177/1073858420902360] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The opioidergic system and intrinsic brain activity, as organized in large-scale networks such as the salience network (SN), sensorimotor network (SMN), and default-mode network (DMN), play core roles in healthy behavior and psychiatric disorders. This work aimed to investigate how opioidergic signaling affects intrinsic brain activity in healthy individuals by reviewing relevant neuroanatomical, molecular, functional, and pharmacological magnetic resonance imaging studies in order to clarify their physiological links and changes in psychiatric disorders. The SN shows dense opioidergic innervations of subcortical structures and high expression levels of opioid receptors in subcortical-cortical areas, with enhanced or reduced activity with low or very high doses of opioids, respectively. The SMN shows high levels of opioid receptors in subcortical areas and functional disconnection caused by opioids. The DMN shows low levels of opioid receptors in cortical areas and inhibited or enhanced activity with low or high doses of opioids, respectively. Finally, we proposed a working model. Opioidergic signaling enhances SN and suppresses SMN (and DMN) activity, resulting in affective excitation with psychomotor inhibition; stronger increases in opioidergic signaling attenuate the SN and SMN while disinhibiting the DMN, dissociating affective and psychomotor functions from the internal states; the opposite occurs with a deficit of opioidergic signaling.
Collapse
Affiliation(s)
- Giulio Rocchi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Bruno Sterlini
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Samuele Tardito
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Matilde Inglese
- Ospedale Policlinico San Martino IRCCS, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy
| | - Anna Corradi
- Ospedale Policlinico San Martino IRCCS, Genoa, Italy
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Gilberto Filaci
- Ospedale Policlinico San Martino IRCCS, Genoa, Italy
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Paola Magioncalda
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy
- Brain and Consciousness Research Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan
| | - Matteo Martino
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
16
|
McCluskey SP, Plisson C, Rabiner EA, Howes O. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. Eur J Nucl Med Mol Imaging 2020; 47:451-489. [PMID: 31541283 PMCID: PMC6974496 DOI: 10.1007/s00259-019-04488-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE A limit on developing new treatments for a number of central nervous system (CNS) disorders has been the inadequate understanding of the in vivo pathophysiology underlying neurological and psychiatric disorders and the lack of in vivo tools to determine brain penetrance, target engagement, and relevant molecular activity of novel drugs. Molecular neuroimaging provides the tools to address this. This article aims to provide a state-of-the-art review of new PET tracers for CNS targets, focusing on developments in the last 5 years for targets recently available for in-human imaging. METHODS We provide an overview of the criteria used to evaluate PET tracers. We then used the National Institute of Mental Health Research Priorities list to identify the key CNS targets. We conducted a PubMed search (search period 1st of January 2013 to 31st of December 2018), which yielded 40 new PET tracers across 16 CNS targets which met our selectivity criteria. For each tracer, we summarised the evidence of its properties and potential for use in studies of CNS pathophysiology and drug evaluation, including its target selectivity and affinity, inter and intra-subject variability, and pharmacokinetic parameters. We also consider its potential limitations and missing characterisation data, but not specific applications in drug development. Where multiple tracers were present for a target, we provide a comparison of their properties. RESULTS AND CONCLUSIONS Our review shows that multiple new tracers have been developed for proteinopathy targets, particularly tau, as well as the purinoceptor P2X7, phosphodiesterase enzyme PDE10A, and synaptic vesicle glycoprotein 2A (SV2A), amongst others. Some of the most promising of these include 18F-MK-6240 for tau imaging, 11C-UCB-J for imaging SV2A, 11C-CURB and 11C-MK-3168 for characterisation of fatty acid amide hydrolase, 18F-FIMX for metabotropic glutamate receptor 1, and 18F-MNI-444 for imaging adenosine 2A. Our review also identifies recurrent issues within the field. Many of the tracers discussed lack in vivo blocking data, reducing confidence in selectivity. Additionally, late-stage identification of substantial off-target sites for multiple tracers highlights incomplete pre-clinical characterisation prior to translation, as well as human disease state studies carried out without confirmation of test-retest reproducibility.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| | - Christophe Plisson
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
17
|
de Oliveira PG, Ramos MLS, Amaro AJ, Dias RA, Vieira SI. G i/o-Protein Coupled Receptors in the Aging Brain. Front Aging Neurosci 2019; 11:89. [PMID: 31105551 PMCID: PMC6492497 DOI: 10.3389/fnagi.2019.00089] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/03/2019] [Indexed: 12/18/2022] Open
Abstract
Cells translate extracellular signals to regulate processes such as differentiation, metabolism and proliferation, via transmembranar receptors. G protein-coupled receptors (GPCRs) belong to the largest family of transmembrane receptors, with over 800 members in the human species. Given the variety of key physiological functions regulated by GPCRs, these are main targets of existing drugs. During normal aging, alterations in the expression and activity of GPCRs have been observed. The central nervous system (CNS) is particularly affected by these alterations, which results in decreased brain functions, impaired neuroregeneration, and increased vulnerability to neuropathologies, such as Alzheimer's and Parkinson diseases. GPCRs signal via heterotrimeric G proteins, such as Go, the most abundant heterotrimeric G protein in CNS. We here review age-induced effects of GPCR signaling via the Gi/o subfamily at the CNS. During the aging process, a reduction in protein density is observed for almost half of the Gi/o-coupled GPCRs, particularly in age-vulnerable regions such as the frontal cortex, hippocampus, substantia nigra and striatum. Gi/o levels also tend to decrease with aging, particularly in regions such as the frontal cortex. Alterations in the expression and activity of GPCRs and coupled G proteins result from altered proteostasis, peroxidation of membranar lipids and age-associated neuronal degeneration and death, and have impact on aging hallmarks and age-related neuropathologies. Further, due to oligomerization of GPCRs at the membrane and their cooperative signaling, down-regulation of a specific Gi/o-coupled GPCR may affect signaling and drug targeting of other types/subtypes of GPCRs with which it dimerizes. Gi/o-coupled GPCRs receptorsomes are thus the focus of more effective therapeutic drugs aiming to prevent or revert the decline in brain functions and increased risk of neuropathologies at advanced ages.
Collapse
Affiliation(s)
- Patrícia G de Oliveira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - Marta L S Ramos
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - António J Amaro
- School of Health Sciences (ESSUA), Universidade de Aveiro, Aveiro, Portugal
| | - Roberto A Dias
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - Sandra I Vieira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| |
Collapse
|
18
|
Li S, Zheng MQ, Naganawa M, Gao H, Pracitto R, Shirali A, Lin SF, Teng JK, Ropchan J, Huang Y. Novel Kappa Opioid Receptor Agonist as Improved PET Radiotracer: Development and in Vivo Evaluation. Mol Pharm 2019; 16:1523-1531. [PMID: 30726092 DOI: 10.1021/acs.molpharmaceut.8b01209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The kappa opioid receptor (KOR) is involved in depression, alcoholism, and drug abuse. The current agonist radiotracer 11C-GR103545 is not ideal for imaging KOR due to its slow tissue kinetics in human. The aim of our project was to develop novel KOR agonist radiotracers with improved imaging properties. A novel compound FEKAP ((( R))-4-(2-(3,4-dichlorophenyl)acetyl)-3-((ethyl(2-fluoroethyl)amino)methyl) piperazine-1-carboxylate) was designed, synthesized, and assayed for in vitro binding affinities. It was then radiolabeled and evaluated in rhesus monkeys. Baseline and blocking scans were conducted on a Focus-220 scanner to assess binding specificity and selectivity. Metabolite-corrected arterial activities over time were measured and used as input functions to analyze the brain regional time-activity curves and derive kinetic and binding parameters with kinetic modeling. FEKAP displayed high KOR binding affinity ( Ki = 0.43 nM) and selectivity (17-fold over mu opioid receptor and 323-fold over delta opioid receptor) in vitro. 11C-FEKAP was prepared in high molar activity (mean of 718 GBq/μmol, n = 19) and >99% radiochemical purity. In monkeys, 11C-FEKAP metabolized fairly fast, with ∼31% of intact parent fraction at 30 min post-injection. In the brain, it exhibited fast and reversible kinetics with good uptake. Pretreatment with the nonselective opioid receptor antagonist naloxone (1 mg/kg) decreased uptake in high binding regions to the level in the cerebellum, and the selective KOR antagonist LY2456302 (0.02 and 0.1 mg/kg) reduced 11C-FEKAP specific binding in a dose-dependent manner. As a measure of specific binding signals, the mean binding potential ( BPND) values of 11C-FEKAP derived from the multilinear analysis-1 (MA1) method were greater than 0.5 for all regions, except for the thalamus. The novel KOR agonist tracer 11C-FEKAP demonstrated binding specificity and selectivity in vivo and exhibited attractive properties of fast tissue kinetics and high specific binding.
Collapse
Affiliation(s)
- Songye Li
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Ming-Qiang Zheng
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Mika Naganawa
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Hong Gao
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Richard Pracitto
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Anupama Shirali
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Shu-Fei Lin
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Jo-Ku Teng
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Jim Ropchan
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| |
Collapse
|
19
|
Guerrero M, Urbano M, Kim EK, Gamo AM, Riley S, Abgaryan L, Leaf N, Van Orden LJ, Brown SJ, Xie JY, Porreca F, Cameron MD, Rosen H, Roberts E. Design and Synthesis of a Novel and Selective Kappa Opioid Receptor (KOR) Antagonist (BTRX-335140). J Med Chem 2019; 62:1761-1780. [PMID: 30707578 PMCID: PMC6395531 DOI: 10.1021/acs.jmedchem.8b01679] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
κ opioid receptor (KOR) antagonists are potential pharmacotherapies for the treatment of migraine and stress-related mood disorders including depression, anxiety, and drug abuse, thus the development of novel KOR antagonists with an improved potency/selectivity profile and medication-like duration of action has attracted the interest of the medicinal chemistry community. In this paper, we describe the discovery of 1-(6-ethyl-8-fluoro-4-methyl-3-(3-methyl-1,2,4-oxadiazol-5-yl)quinolin-2-yl)- N-(tetrahydro-2 H-pyran-4-yl)piperidin-4 amine (CYM-53093, BTRX-335140) as a potent and selective KOR antagonist, endowed with favorable in vitro ADMET and in vivo pharmacokinetic profiles and medication-like duration of action in rat pharmacodynamic experiments. Orally administered CYM-53093 showed robust efficacy in antagonizing KOR agonist-induced prolactin secretion and in tail-flick analgesia in mice. CYM-53093 exhibited a broad selectivity over a panel of off-target proteins. This compound is in phase 1 clinical trials for the treatment of neuropsychiatric disorders wherein dynorphin is thought to contribute to the underlying pathophysiology.
Collapse
Affiliation(s)
- Miguel Guerrero
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Mariangela Urbano
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Eun-Kyong Kim
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Ana M. Gamo
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Sean Riley
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Lusine Abgaryan
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Nora Leaf
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Lori Jean Van Orden
- BlackThorn Therapeutics, Inc. 780 Brannan St. San Francisco CA 94103, United States
| | - Steven J. Brown
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Jennifer Y. Xie
- Current address: Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine @ Arkansas State University, Jonesboro, AR 72446, United States
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, United States
| | - Frank Porreca
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, United States
| | - Michael D. Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Hugh Rosen
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Edward Roberts
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| |
Collapse
|
20
|
Usai EM, Manca I, Pettinau F, Mastino A, Pittau B. Chemical Characterization and in vitro
Metabolism of a Novel Class of Delta Opioid Receptor Agonists, Analogs of SNC-80. ChemistrySelect 2019. [DOI: 10.1002/slct.201803906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Elisabetta Maria Usai
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| | - Ilaria Manca
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| | - Francesca Pettinau
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| | - Antonio Mastino
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
- Department of Chemical; Biological, Pharmaceutical, and Environmental Sciences; University of Messina; Messina Italy
| | - Barbara Pittau
- Institute of Translational Pharmacology; National Research Council; 09010 Pula (CA) Italy
| |
Collapse
|
21
|
Torres-Berrio A, Nava-Mesa MO. The opioid system in stress-induced memory disorders: From basic mechanisms to clinical implications in post-traumatic stress disorder and Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:327-338. [PMID: 30118823 DOI: 10.1016/j.pnpbp.2018.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/25/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
Cognitive and emotional impairment are a serious consequence of stress exposure and are core features of neurological and psychiatric conditions that involve memory disorders. Indeed, acute and chronic stress are high-risk factors for the onset of post-traumatic stress disorder (PTSD) and Alzheimer's disease (AD), two devastating brain disorders associated with memory dysfunction. Besides the sympathetic nervous system and the hypothalamic-pituitary-adrenal (HPA) axis, stress response also involves the activation of the opioid system in brain regions associated with stress regulation and memory processing. In this context, it is possible that stress-induced memory disorders may be attributed to alterations in the interaction between the neuroendocrine stress system and the opioid system. In this review, we: (1) describe the effects of acute and chronic stress on memory, and the modulatory role of the opioid system, (2) discuss the contribution of the opioid system to the pathophysiology of PTSD and AD, and (3) present evidence of current and potential therapies that target the opioid receptors to treat PTSD- and AD-associated symptoms.
Collapse
Affiliation(s)
| | - Mauricio O Nava-Mesa
- Neuroscience Research Group (NEUROS), School of Medicine, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
22
|
Analysis of natural product regulation of opioid receptors in the treatment of human disease. Pharmacol Ther 2018; 184:51-80. [DOI: 10.1016/j.pharmthera.2017.10.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
23
|
Butelman ER, Kreek MJ. Discriminative Stimulus Properties of Opioid Ligands: Progress and Future Directions. Curr Top Behav Neurosci 2018; 39:175-192. [PMID: 27225498 DOI: 10.1007/7854_2016_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Opioid receptors (MOP-r, KOP-r, DOP-r, as well as NOP-r) and their endogenous neuropeptide agonist systems are involved in diverse neurobiological and behavioral functions, in health and disease. These functions include pain and analgesia, addictions, and psychiatric diseases (e.g., depression-, anxiety-like, and stress-related disorders). Drug discrimination assays have been used to characterize the behavioral pharmacology of ligands with affinity at MOP-r, KOP-r, or DOP-r (and to a lesser extent NOP-r). Therefore, drug discrimination studies with opioid ligands have an important continuing role in translational investigations of diseases that are affected by these neurobiological targets and their pharmacotherapy.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY, 10065, USA.
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY, 10065, USA
| |
Collapse
|
24
|
Burtscher J, Schwarzer C. The Opioid System in Temporal Lobe Epilepsy: Functional Role and Therapeutic Potential. Front Mol Neurosci 2017; 10:245. [PMID: 28824375 PMCID: PMC5545604 DOI: 10.3389/fnmol.2017.00245] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/24/2017] [Indexed: 12/13/2022] Open
Abstract
Temporal lobe epilepsy is considered to be one of the most common and severe forms of focal epilepsies. Patients often develop cognitive deficits and emotional blunting along the progression of the disease. The high incidence of resistance to antiepileptic drugs and a frequent lack of admissibility to surgery poses an unmet medical challenge. In the urgent quest of novel treatment strategies, neuropeptides are interesting candidates, however, their therapeutic potential has not yet been exploited. This review focuses on the functional role of the endogenous opioid system with respect to temporal lobe epilepsy, specifically in the hippocampus. The role of dynorphins and kappa opioid receptors (KOPr) as modulators of neuronal excitability is well understood: both the reduced release of glutamate as well of postsynaptic hyperpolarization were shown in glutamatergic neurons. In line with this, low levels of dynorphin in humans and mice increase the risk of epilepsy development. The role of enkephalins is not understood so well. On one hand, some agonists of the delta opioid receptors (DOPr) display pro-convulsant properties probably through inhibition of GABAergic interneurons. On the other hand, enkephalins play a neuro-protective role under hypoxic or anoxic conditions, most probably through positive effects on mitochondrial function. Despite the supposed absence of endorphins in the hippocampus, exogenous activation of the mu opioid receptors (MOPr) induces pro-convulsant effects. Recently-expanded knowledge of the complex ways opioid receptors ligands elicit their effects (including biased agonism, mixed binding, and opioid receptor heteromers), opens up exciting new therapeutic potentials with regards to seizures and epilepsy. Potential adverse side effects of KOPr agonists may be minimized through functional selectivity. Preclinical data suggest a high potential of such compounds to control seizures, with a strong predictive validity toward human patients. The discovery of DOPr-agonists without proconvulsant potential stimulates the research on the therapeutic use of neuroprotective potential of the enkephalin/DOPr system.
Collapse
Affiliation(s)
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University of InnsbruckInnsbruck, Austria
| |
Collapse
|
25
|
Raina R, Sen D. Can crosstalk between DOR and PARP reduce oxidative stress mediated neurodegeneration? Neurochem Int 2017; 112:206-218. [PMID: 28739183 DOI: 10.1016/j.neuint.2017.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/15/2017] [Accepted: 07/19/2017] [Indexed: 12/20/2022]
Abstract
The progressive loss of structure and function of neurons leads to neurodegenerative processes which become the causative reason for various neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD) etc. These diseases are multifactorial in nature but they have been seen to possess similar causative agents to a certain extent. Oxidative Stress (OS) has been identified as a major stressor and a mediator in most of these diseases. OS not only leads to the generation of free radical species but if persistent, can possibly lead to lipid peroxidation, protein damage, DNA damage, and cell death. Anti-oxidants are endogenously present in our body to tackle oxygen metabolites but their levels reduce greatly under continuous OS conditions. In such a case, dietary supplements to replenish the anti-oxidant levels in our body is a good way of treatment but it is very slow and may not be as effective in chronic stress conditions. Thus, there is a need for more effective mechanisms to attenuate OS. Two such mechanisms which can be considered are the activation of Delta opioid receptor (DOR) and Inhibition of Poly (ADP-ribose)-polymerase1 (PARP1), which have been suggested to protect neurons and increase neuronal cell survivability in both in-vitro and in-vivo disease models. Various signaling pathways have been highlighted to probably play a significant role in attenuating OS by the activation of DOR. It would be an interesting topic of investigation to see if one of the probable mechanisms by which DOR attenuates OS could be by modulation of PARP through a cascade of intracellular signaling reactions.
Collapse
Affiliation(s)
- Rutika Raina
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), VIT University, Vellore, Tamil Nadu, India
| | - Dwaipayan Sen
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), VIT University, Vellore, Tamil Nadu, India.
| |
Collapse
|
26
|
Lesniak A, Leszczynski P, Bujalska-Zadrozny M, Pick CG, Sacharczuk M. Naloxone exacerbates memory impairments and depressive-like behavior after mild traumatic brain injury (mTBI) in mice with upregulated opioid system activity. Behav Brain Res 2017; 326:209-216. [PMID: 28284950 DOI: 10.1016/j.bbr.2017.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/02/2017] [Accepted: 03/07/2017] [Indexed: 12/13/2022]
Abstract
The neuroprotective role of the endogenous opioid system in the pathophysiological sequelae of brain injury remains largely ambiguous. Noteworthy, almost no data is available on how its genetically determined activity influences the outcome of mild traumatic brain injury. Thus, the aim of our study was to examine the effect of opioid receptor blockage on cognitive impairments produced by mild traumatic brain injury in mice selectively bred for high (HA) and low (LA) swim-stress induced analgesia that show innate divergence in opioid system activity. Mild traumatic brain injury was induced with a weight-drop device on anaesthetized mice. Naloxone (5mg/kg) was intraperitoneally delivered twice a day for 7days to non-selectively block opioid receptors. Spatial memory performance and manifestations of depressive-like behavior were assessed using the Morris Water Maze and tail suspension tests, respectively. Mild traumatic brain injury resulted in a significant deterioration of spatial memory performance and severity of depressive-like behavior in the LA mouse line as opposed to HA mice. Opioid receptor blockage with naloxone unmasked cognitive deficits in HA mice but was without effect in the LA line. The results suggest a protective role of genetically predetermined enhanced opioid system activity in suppression of mild brain trauma-induced cognitive impairments. Mice selected for high and low swim stress-induced analgesia might therefore be a useful model to study the involvement of the opioid system in the pathophysiology and neurological outcome of traumatic brain injury.
Collapse
Affiliation(s)
- Anna Lesniak
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Centre for Preclinical Research and Technology (CePT), Banacha 1B, 02-097 Warsaw, Poland
| | - Pawel Leszczynski
- Department of Genomics, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, Postepu 36A, 05-552 Magdalenka, Poland
| | - Magdalena Bujalska-Zadrozny
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Centre for Preclinical Research and Technology (CePT), Banacha 1B, 02-097 Warsaw, Poland
| | - Chaim G Pick
- Department of Anatomy, and Anthropology, Sackler Faculty of Medicine, and Sagol School of Neuroscience, Tel Aviv University, Ramat-Aviv, 69978 Tel Aviv, Israel
| | - Mariusz Sacharczuk
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Centre for Preclinical Research and Technology (CePT), Banacha 1B, 02-097 Warsaw, Poland; Department of Internal Medicine, Hypertension and Vascular Diseases, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; Department of Genomics, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, Postepu 36A, 05-552 Magdalenka, Poland.
| |
Collapse
|
27
|
Ji H, Wang Y, Liu G, Chang L, Chen Z, Zhou D, Xu X, Cui W, Hong Q, Jiang L, Li J, Zhou X, Li Y, Guo Z, Zha Q, Niu Y, Weng Q, Duan S, Wang Q. Elevated OPRD1 promoter methylation in Alzheimer's disease patients. PLoS One 2017; 12:e0172335. [PMID: 28253273 PMCID: PMC5333823 DOI: 10.1371/journal.pone.0172335] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/03/2017] [Indexed: 01/21/2023] Open
Abstract
Aberrant DNA methylation has been observed in the patients with Alzheimer’s disease (AD), a common neurodegenerative disorder in the elderly. OPRD1 encodes the delta opioid receptor, a member of the opioid family of G-protein-coupled receptors. In the current study, we compare the DNA methylation levels of OPRD1 promoter CpG sites (CpG1, CpG2, and CpG3) between 51 AD cases and 63 controls using the bisulfite pyrosequencing technology. Our results show that significantly higher CpG3 methylation is found in AD cases than controls. Significant associations are found between several biochemical parameters (including HDL-C and ALP) and CpG3 methylation. Subsequent luciferase reporter gene assay shows that DNA fragment containing the three OPRD1 promoter CpGs is able to regulate gene expression. In summary, our results suggest that OPRD1 promoter hypermethylation is associated with the risk of AD.
Collapse
Affiliation(s)
- Huihui Ji
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yunliang Wang
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, Shandong, China
| | - Guili Liu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Lan Chang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | | | | | - Xuting Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Wei Cui
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Qingxiao Hong
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Liting Jiang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Jinfeng Li
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, Shandong, China
| | - Xiaohui Zhou
- Department of Internal Medicine for Cadres, the First Affiliated Hospital of Xinjiang Medical University, Urumchi, China
| | - Ying Li
- Ningbo No. 1 Hospital, Ningbo, Zhejiang, China
| | - Zhiping Guo
- School of Medicine, Lishui University, Lishui, Zhejiang, China
| | - Qin Zha
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (QW); (SD); (QZ)
| | - Yanfang Niu
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qiuyan Weng
- The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shiwei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (QW); (SD); (QZ)
| | - Qinwen Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (QW); (SD); (QZ)
| |
Collapse
|
28
|
Hermann D, Hirth N, Reimold M, Batra A, Smolka MN, Hoffmann S, Kiefer F, Noori HR, Sommer WH, Reischl G, la Fougère C, Mann K, Spanagel R, Hansson AC. Low μ-Opioid Receptor Status in Alcohol Dependence Identified by Combined Positron Emission Tomography and Post-Mortem Brain Analysis. Neuropsychopharmacology 2017; 42:606-614. [PMID: 27510425 PMCID: PMC5240173 DOI: 10.1038/npp.2016.145] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/20/2016] [Accepted: 07/22/2016] [Indexed: 12/20/2022]
Abstract
Blockade of the μ-opioid receptor (MOR) by naltrexone reduces relapse risk in a subpopulation of alcohol-dependent patients. Previous positron-emission-tomography (PET) studies using the MOR ligand [11C]carfentanil have found increased MOR availability in abstinent alcoholics, which may reflect either increased MOR expression or lower endogenous ligand concentration. To differentiate between both effects, we investigated two cohorts of alcoholic subjects using either post-mortem or clinical PET analysis. Post-mortem brain tissue of alcohol-dependent subjects and controls (N=43/group) was quantitatively analyzed for MOR ([3H]DAMGO)-binding sites and OPRM1 mRNA in striatal regions. [11C]carfentanil PET was performed in detoxified, medication free alcohol-dependent patients (N=38), followed by a randomized controlled study of naltrexone versus placebo and follow-up for 1 year (clinical trial number: NCT00317031). Because the functional OPRM1 variant rs1799971:A>G affects the ligand binding, allele carrier status was considered in the analyses. MOR-binding sites were reduced by 23-51% in post-mortem striatal tissue of alcoholics. In the PET study, a significant interaction of OPRM1 genotype, binding potential (BPND) for [11C]carfentanil in the ventral striatum, and relapse risk was found. Particularly in G-allele carriers, lower striatal BPND was associated with a higher relapse risk. Interestingly, this effect was more pronounced in the naltrexone treatment group. Reduced MOR is interpreted as a neuroadaptation to an alcohol-induced release of endogenous ligands in patients with severe alcoholism. Low MOR availability may explain the ineffectiveness of naltrexone treatment in this subpopulation. Finally, low MOR-binding sites are proposed as a molecular marker for a negative disease course.
Collapse
Affiliation(s)
- Derik Hermann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,Central Institute of Mental Health (ZI), Square J5, Mannheim 68159, Germany, Tel: +49 621 1703 6293 or +49 621 1703 3522, Fax: 49 621 17036255,E-mail: or
| | - Natalie Hirth
- Institute of Psychopharmacology, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Reimold
- Department of Nuclear Medicine, University of Tübingen, Tübingen, Germany
| | - Anil Batra
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Michael N Smolka
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Sabine Hoffmann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hamid R Noori
- Institute of Psychopharmacology, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolfgang H Sommer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,Institute of Psychopharmacology, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | | | - Karl Mann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anita C Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,Central Institute of Mental Health (ZI), Square J5, Mannheim 68159, Germany, Tel: +49 621 1703 6293 or +49 621 1703 3522, Fax: 49 621 17036255,E-mail: or
| |
Collapse
|
29
|
Cai Z, Li S, Pracitto R, Navarro A, Shirali A, Ropchan J, Huang Y. Fluorine-18-Labeled Antagonist for PET Imaging of Kappa Opioid Receptors. ACS Chem Neurosci 2017; 8:12-16. [PMID: 27741398 DOI: 10.1021/acschemneuro.6b00268] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Kappa opioid receptor (KOR) antagonists are potential drug candidates for diseases such as treatment-refractory depression, anxiety, and addictive disorders. PET imaging radiotracers for KOR can be used in occupancy study to facilitate drug development, and to investigate the roles of KOR in health and diseases. We have previously developed two 11C-labeled antagonist radiotracers with high affinity and selectivity toward KOR. What is limiting their wide applications is the short half-life of 11C. Herein, we report the synthesis of a first 18F-labeled KOR antagonist radiotracer and the initial PET imaging study in a nonhuman primate.
Collapse
Affiliation(s)
- Zhengxin Cai
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Songye Li
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Richard Pracitto
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Antonio Navarro
- Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Anupama Shirali
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Jim Ropchan
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Yiyun Huang
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| |
Collapse
|
30
|
Liu G, Ji H, Liu J, Xu C, Chang L, Cui W, Ye C, Hu H, Chen Y, Zhou X, Duan S, Wang Q. Association of OPRK1 and OPRM1 methylation with mild cognitive impairment in Xinjiang Han and Uygur populations. Neurosci Lett 2017; 636:170-176. [DOI: 10.1016/j.neulet.2016.11.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/13/2016] [Accepted: 11/07/2016] [Indexed: 12/01/2022]
|
31
|
Crist RC, Clarke TK. OPRD1 Genetic Variation and Human Disease. Handb Exp Pharmacol 2016; 247:131-145. [PMID: 28035534 DOI: 10.1007/164_2016_112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The OPRD1 gene encodes the delta-opioid receptor, which has multiple functions including regulating reward pathways. The gene contains more than 2,000 verified genetic variants but only 2 currently have evidence for specific functions: rs1042114 disrupts maturation of the receptor and rs569356 affects OPRD1 expression. These polymorphisms and others in the gene have been found to be associated with human diseases. The most reproducible data are associations between opioid addiction and three variants in intron 1 (rs2236861, rs2236857, and rs3766951), which have been described in a number of independent populations. Several publications also point toward an association between anorexia and a haplotype block containing rs569356 and rs533123. Unfortunately the mechanisms underlying these two effects are currently unknown. In contrast, rs1042114 has been linked to Alzheimer's disease through an increasingly well-defined mechanism by which the variant allele reduces production of the beta-amyloid plaques associated with the disease. Additional studies of OPRD1 variants are necessary to replicate current findings and to delineate the functional roles of relevant polymorphisms.
Collapse
Affiliation(s)
- Richard C Crist
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, 125 South 31st Street, Room 2207, Philadelphia, PA, 19104, USA.
| | - Toni-Kim Clarke
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| |
Collapse
|
32
|
Schmitt S, Delamare J, Tirel O, Fillesoye F, Dhilly M, Perrio C. N-[ 18F]-FluoropropylJDTic for κ-opioid receptor PET imaging: Radiosynthesis, pre-clinical evaluation, and metabolic investigation in comparison with parent JDTic. Nucl Med Biol 2016; 44:50-61. [PMID: 27821345 DOI: 10.1016/j.nucmedbio.2016.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/07/2016] [Accepted: 09/17/2016] [Indexed: 02/08/2023]
Abstract
INTRODUCTION To image kappa opioid receptor (KOR) for preclinical studies, N-fluoropropylJDTic 9 derived from the best-established KOR antagonist JDTic, was labeled with fluorine-18. METHODS Radiosynthesis of [18F]9 was achieved according to an automated two-step procedure from [18F]-fluoride. Peripheral and cerebral distributions were determined by ex vivo experiments and by PET imaging in mouse. Radiometabolism studies were performed both in vivo in mice and in vitro in mouse and human liver microsomes. Identification of the major metabolic fragmentations was carried out by UPLC-MS analysis of enzymatic cleavage of non-radioactive ligand 9. Microsomal metabolic degradation of parent JDTic was also achieved for comparison. RESULTS The radiotracer [18F]9 was produced after 140±5min total synthesis time (2.2±0.4% not decay corrected radiochemical yield) with a specific activity of 41-89GBq/μmol (1.1-2.4Ci/μmol). Peripheral and regional brain distributions of [18F]9 were consistent with known KOR locations but no significant specific binding in brain was shown. [18F]9 presented a typical hepatobiliary and renal elimination, and was rapidly metabolized. The in vivo and in vitro radiometabolic profiles of [18F]9 were similar. Piperidine 12 was identified as the major metabolic fragment of the non-radioactive ligand 9. JDTic 7 was found to be much more stable than 9. CONCLUSION Although the newly proposed radioligand [18F]9 was concluded to be not suitable for KOR PET imaging due to the formation of brain penetrating radiometabolites, our findings highlight the metabolic stability of JDTic and may help in the design of novel JDTic derivatives for in vivo applications.
Collapse
Affiliation(s)
- Sébastien Schmitt
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Jérôme Delamare
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Olivier Tirel
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Fabien Fillesoye
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Martine Dhilly
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Cécile Perrio
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France.
| |
Collapse
|
33
|
Fluegge K. Does MeCP2 deficiency in autism confer protection against later development of Alzheimer’s disease? A reply to Oberman and Pascual-Leone (2014). Med Hypotheses 2016; 92:18-20. [DOI: 10.1016/j.mehy.2016.04.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 02/08/2023]
|
34
|
Butelman ER, Kreek MJ. Salvinorin A, a kappa-opioid receptor agonist hallucinogen: pharmacology and potential template for novel pharmacotherapeutic agents in neuropsychiatric disorders. Front Pharmacol 2015; 6:190. [PMID: 26441647 PMCID: PMC4561799 DOI: 10.3389/fphar.2015.00190] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/24/2015] [Indexed: 12/19/2022] Open
Abstract
Salvinorin A is a potent hallucinogen, isolated from the ethnomedical plant Salvia divinorum. Salvinorin A is a selective high efficacy kappa-opioid receptor (KOPr) agonist, and thus implicates the KOPr system and its endogenous agonist ligands (the dynorphins) in higher functions, including cognition and perceptual effects. Salvinorin A is the only selective KOPr ligand to be widely available outside research or medical settings, and salvinorin A-containing products have undergone frequent non-medical use. KOPr/dynorphin systems in the brain are known to be powerful counter-modulatory mechanisms to dopaminergic function, which is important in mood and reward engendered by natural and chemical reinforcers (including drugs of abuse). KOPr activation (including by salvinorin A) can thus cause aversion and anhedonia in preclinical models. Salvinorin A is also a completely new scaffold for medicinal chemistry approaches, since it is a non-nitrogenous neoclerodane, unlike other known opioid ligands. Ongoing efforts have the goal of discovering novel semi-synthetic salvinorin analogs with potential KOPr-mediated pharmacotherapeutic effects (including partial agonist or biased agonist effects), with a reduced burden of undesirable effects associated with salvinorin A.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University , New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University , New York, NY, USA
| |
Collapse
|
35
|
Ji H, Wang Y, Liu G, Xu X, Dai D, Chen Z, Zhou D, Zhou X, Han L, Li Y, Zhuo R, Hong Q, Jiang L, Zhang X, Liu Y, Xu L, Chang L, Li J, An P, Duan S, Wang Q. OPRK1 promoter hypermethylation increases the risk of Alzheimer's disease. Neurosci Lett 2015; 606:24-9. [PMID: 26300544 DOI: 10.1016/j.neulet.2015.08.027] [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: 12/31/2014] [Revised: 08/07/2015] [Accepted: 08/16/2015] [Indexed: 10/23/2022]
Abstract
As a member of the opioid family, κ-opioid receptors play important role in cognitive and learning functions. The purpose of this study was to evaluate the association of OPRK1 promoter methylation with Alzheimer's disease (AD). OPRK1 DNA methylation levels of 48 cases and 58 well matched controls were measured using the bisulphite pyrosequencing technology. Our results showed that there was a significant correlation between three CpG sites on the OPRK1 promoter region (r>=0.715, p<0.001). Thus, the mean methylation value of the three CpG sites was used for the case-control comparison. And our results showed there was a significantly higher OPRK1 promoter methylation in AD cases than in controls (p=0.006, adjusted p=0.012). Subsequent luciferase reporter assay showed the CpGs containing fragment of OPRK1 promoter significantly increased the expression of reporter gene (Fold=2.248, p=0.0235). In summary, our results suggested that OPRK1 promoter hypermethylation might increase the risk of AD through its regulation on the gene expression of OPRK1.
Collapse
Affiliation(s)
- Huihui Ji
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yunliang Wang
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, Shandong 255000, China.
| | - Guili Liu
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xuting Xu
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Dongjun Dai
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | | | | | - Xiaohui Zhou
- Department of Internal Medicine for Cadres, the First Affiliated Hospital of Xinjiang Medical University, Urumchi 830000, China
| | - Liyuan Han
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Ying Li
- Ningbo No. 1 Hospital, Zhejiang 315200, China
| | - Renjie Zhuo
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Qingxiao Hong
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Liting Jiang
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xiaonan Zhang
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yu Liu
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Lei Xu
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Lan Chang
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jinfeng Li
- Department of Neurology, the 148 Central Hospital of PLA, Zibo, Shandong 255000, China
| | - Pengyuan An
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Shiwei Duan
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Qinwen Wang
- Ningbo Key Lab of Behavior Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
36
|
Laukkanen V, Kärkkäinen O, Kautiainen H, Tiihonen J, Storvik M. Decreased [³H]naloxone Binding in the Dentate Gyrus of Cloninger Type 1 Anxiety-Prone Alcoholics: A Postmortem Whole-Hemisphere Autoradiography Study. Alcohol Clin Exp Res 2015; 39:1352-9. [DOI: 10.1111/acer.12791] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/26/2015] [Indexed: 01/20/2023]
Affiliation(s)
- Virpi Laukkanen
- Department of Forensic Psychiatry; University of Eastern Finland; Niuvanniemi Hospital; Kuopio Finland
- Department of Psychiatry; Kuopio University Hospital; Kuopio Finland
| | - Olli Kärkkäinen
- Department of Pharmacology and Toxicology; University of Eastern Finland; Kuopio Finland
| | - Hannu Kautiainen
- Unit of Primary Health Care; Helsinki University Central Hospital; Helsinki Finland
- Department of General Practice; University of Helsinki; Helsinki Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry; University of Eastern Finland; Niuvanniemi Hospital; Kuopio Finland
- Department of Clinical Neuroscience; Karolinska Institutet; Karolinska sjukhuset; Stockholm Sweden
| | - Markus Storvik
- Department of Pharmacology and Toxicology; University of Eastern Finland; Kuopio Finland
| |
Collapse
|
37
|
Chang RYK, Etheridge N, Nouwens AS, Dodd PR. SWATH analysis of the synaptic proteome in Alzheimer's disease. Neurochem Int 2015; 87:1-12. [PMID: 25958317 DOI: 10.1016/j.neuint.2015.04.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/20/2015] [Accepted: 04/21/2015] [Indexed: 10/23/2022]
Abstract
Brain tissue from Alzheimer's disease patients exhibits synaptic degeneration in selected regions. Synaptic dysfunction occurs early in the disease and is a primary pathological target for treatment. The molecular mechanisms underlying this degeneration remain unknown. Quantifying the synaptic proteome in autopsy brain and comparing tissue from Alzheimer's disease cases and subjects with normal aging are critical to understanding the molecular mechanisms associated with Alzheimer pathology. We isolated synaptosomes from hippocampus and motor cortex so as to reduce sample complexity relative to whole-tissue homogenates. Synaptosomal extracts were subjected to strong cation exchange (SCX) fractionation to further partition sample complexity; each fraction received SWATH-based information-dependent acquisition to generate a comprehensive peptide-ion library. The expression of synaptic proteins from AD hippocampus and motor cortex was then compared between groups. A total of 2077 unique proteins were identified at a critical local false discovery rate <5%. Thirty of these, including 17 novel proteins, exhibited significant expression differences between cases and controls; these proteins are involved in cellular functions including structural maintenance, signal transduction, autophagy, oxidative stress, and proteasome activity, or they have synaptic-vesicle related or energy-related functions. Differentially expressed proteins were subjected to pathway analysis to identify protein-protein interactions. This revealed that the most perturbed molecular and cellular functions were cellular assembly and organization. Core analysis revealed RhoA signaling to be the top canonical pathway. Network analysis showed that differentially expressed proteins were related to cellular assembly and organization, and cellular function and maintenance. This is the first study to combine SCX fractionation with SWATH analysis. SWATH is a promising new technique that can greatly enhance protein identification in any proteome, and has many other benefits; however, there are limitations yet to be resolved.
Collapse
Affiliation(s)
| | - Naomi Etheridge
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia
| | - Amanda S Nouwens
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia
| | - Peter R Dodd
- School of Chemistry and Molecular Biosciences, University of Queensland, Australia.
| |
Collapse
|
38
|
Naganawa M, Zheng MQ, Henry S, Nabulsi N, Lin SF, Ropchan J, Labaree D, Najafzadeh S, Kapinos M, Tauscher J, Neumeister A, Carson RE, Huang Y. Test-retest reproducibility of binding parameters in humans with 11C-LY2795050, an antagonist PET radiotracer for the κ opioid receptor. J Nucl Med 2015; 56:243-8. [PMID: 25593119 DOI: 10.2967/jnumed.114.147975] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED (11)C-LY2795050 is a new antagonist PET radioligand for the κ opioid receptor (KOR). In this study, we assessed the reproducibility of the binding parameters of (11)C-LY2795050 in healthy human subjects. METHODS Sixteen healthy subjects (11 men and 5 women) underwent 2 separate 90-min PET scans with arterial input function and plasma free fraction (fP) measurements. The 2-tissue-compartment model and multilinear analysis-1 were applied to calculate 5 outcome measures in 14 brain regions: distribution volume (VT), VT normalized by fP (VT/fP), and 3 binding potentials (nondisplaceable binding potential, binding potential relative to total plasma concentration, and binding potential relative to free plasma concentration: BPND, BPP, BPF, respectively). Since KOR is distributed ubiquitously throughout the brain, there are no suitable reference regions. We used a fixed fraction of individual cerebellar VT value (VT,CER) as the nondisplaceable VT (VND) (VND = VT,CER/1.17). The relative and absolute test-retest variability and intraclass correlation coefficient were evaluated for the outcome measures of (11)C-LY2795050. RESULTS The test-retest variability of (11)C-LY2795050 for VT was no more than 10% in any region and was 12% in the amygdala. For binding potential (BPND and BPP), the test-retest variability was good in regions of moderate and high KOR density (BPND > 0.4) and poor in regions of low density. Correction by fP (VT/fP or BPF) did not improve the test-retest performance. CONCLUSION Our results suggest that quantification of (11)C-LY2795050 imaging is reproducible and reliable in regions with moderate and high KOR density. Therefore, we conclude that this first antagonist radiotracer is highly useful for PET studies of KOR.
Collapse
Affiliation(s)
- Mika Naganawa
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ming-Qiang Zheng
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Shannan Henry
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Nabeel Nabulsi
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Shu-Fei Lin
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Jim Ropchan
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - David Labaree
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Soheila Najafzadeh
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Michael Kapinos
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | | | - Alexander Neumeister
- Department of Psychiatry and Radiology, New York University School of Medicine, New York, New York
| | - Richard E Carson
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Yiyun Huang
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
39
|
Schmitt S, Colloc'h N, Perrio C. Novel fluoroalkyl derivatives of selective kappa opioid receptor antagonist JDTic: Design, synthesis, pharmacology and molecular modeling studies. Eur J Med Chem 2014; 90:742-50. [PMID: 25513968 DOI: 10.1016/j.ejmech.2014.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/08/2014] [Accepted: 12/10/2014] [Indexed: 11/17/2022]
Abstract
Novel N- and O-fluoroalkyl derivatives of the highly potent KOR antagonist JDTic were designed and synthesized. Their opioid receptor properties were compared in both in vitro binding assays and modeling approach. All compounds displayed nanomolar affinities for KOR. The fluoropropyl derivatives were more active than their fluoroethyl analogues. N-Fluoroalkylation was preferable to O-alkylation to keep a selective KOR binding. Compared to JDTic, the N-fluoropropyl derivative 2 bound to KOR with an only 4-fold lower affinity and a higher selectivity relative to MOR and DOR [Ki(κ) = 1.6 nM; Ki(μ)/Ki(κ) = 12; Ki(δ)/Ki(κ) = 159 for 2versus Ki(κ) = 0.42 nM; Ki(μ)/Ki(κ) = 9; Ki(δ)/Ki(κ) = 85 for JDTic]. Modeling studies based on the crystal structure of the JDTic/KOR complex revealed that fluorine atom in ligand 2 was involved in specific KOR binding. Ligand 2 was concluded to merit further development for KOR exploration.
Collapse
Affiliation(s)
- Sébastien Schmitt
- CNRS, UMR 6301 ISTCT, LDM-TEP, GIP CYCERON, Boulevard Henri Becquerel, 14074 Caen, France; Université de Caen Basse-Normandie, Normandie Univ., France; CEA, DSV/I2BM, France
| | - Nathalie Colloc'h
- Université de Caen Basse-Normandie, Normandie Univ., France; CEA, DSV/I2BM, France; CNRS, UMR 6301 ISTCT, CERVOxy group, GIP CYCERON, Boulevard Henri Becquerel, 14074 Caen, France
| | - Cécile Perrio
- CNRS, UMR 6301 ISTCT, LDM-TEP, GIP CYCERON, Boulevard Henri Becquerel, 14074 Caen, France; Université de Caen Basse-Normandie, Normandie Univ., France; CEA, DSV/I2BM, France.
| |
Collapse
|
40
|
Kinetic modeling of (11)C-LY2795050, a novel antagonist radiotracer for PET imaging of the kappa opioid receptor in humans. J Cereb Blood Flow Metab 2014; 34:1818-25. [PMID: 25182664 PMCID: PMC4269759 DOI: 10.1038/jcbfm.2014.150] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/15/2014] [Accepted: 07/31/2014] [Indexed: 11/08/2022]
Abstract
(11)C-LY2795050 is a novel kappa opioid receptor (KOR) antagonist tracer for positron emission tomography (PET) imaging. The purpose of this first-in-human study was to determine the optimal kinetic model for analysis of (11)C-LY2795050 imaging data. Sixteen subjects underwent baseline scans and blocking scans after oral naltrexone. Compartmental modeling and multilinear analysis-1 (MA1) were applied using the arterial input functions. Two-tissue compartment model and MA1 were found to be the best models to provide reliable measures of binding parameters. The rank order of (11)C-LY2795050 distribution volume (VT) matched the known regional KOR densities in the human brain. Blocking scans with naltrexone indicated no ideal reference region for (11)C-LY2795050. Three methods for calculation of the nondisplaceable distribution volume (VND) were assessed: (1) individual VND estimated from naltrexone occupancy plots, (2) mean VND across subjects, and (3) a fixed fraction of cerebellum VT. Approach (3) produced the lowest intersubject variability in the calculation of binding potentials (BPND, BPF, and BPP). Therefore, binding potentials of (11)C-LY2795050 can be determined if the specific binding fraction in the cerebellum is presumed to be unchanged by diseases and experimental conditions. In conclusion, results from the present study show the suitability of (11)C-LY2795050 to image and quantify KOR in humans.
Collapse
|
41
|
Zheng MQ, Kim SJ, Holden D, Lin SF, Need A, Rash K, Barth V, Mitch C, Navarro A, Kapinos M, Maloney K, Ropchan J, Carson RE, Huang Y. An Improved Antagonist Radiotracer for the κ-Opioid Receptor: Synthesis and Characterization of (11)C-LY2459989. J Nucl Med 2014; 55:1185-91. [PMID: 24854795 DOI: 10.2967/jnumed.114.138701] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 03/25/2014] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The κ-opioid receptors (KORs) are implicated in several neuropsychiatric diseases and addictive disorders. PET with radioligands provides a means to image the KOR in vivo and investigate its function in health and disease. The purpose of this study was to develop the selective KOR antagonist (11)C-LY2459989 as a PET radioligand and characterize its imaging performance in nonhuman primates. METHODS LY2459989 was synthesized and assayed for in vitro binding to opioid receptors. Ex vivo studies in rodents were conducted to assess its potential as a tracer candidate. (11)C-LY2459989 was synthesized by reaction of its iodophenyl precursor with (11)C-cyanide, followed by partial hydrolysis of the resulting (11)C-cyanophenyl intermediate. Imaging experiments with (11)C-LY2459989 were performed in rhesus monkeys with arterial input function measurement. Imaging data were analyzed with kinetic models to derive in vivo binding parameters. RESULTS LY2459989 is a full antagonist with high binding affinity and selectivity for KOR (0.18, 7.68, and 91.3 nM, respectively, for κ, μ, and δ receptors). Ex vivo studies in rats indicated LY2459989 as an appropriate tracer candidate with high specific binding signals and confirmed its KOR binding selectivity in vivo. (11)C-LY2459989 was synthesized in high radiochemical purity and good specific activity. In rhesus monkeys, (11)C-LY2459989 displayed a fast rate of peripheral metabolism. Similarly, (11)C-LY2459989 displayed fast uptake kinetics in the brain and an uptake pattern consistent with the distribution of KOR in primates. Pretreatment with naloxone (1 mg/kg, intravenously) resulted in a uniform distribution of radioactivity in the brain. Further, specific binding of (11)C-LY2459989 was dose-dependently reduced by the selective KOR antagonist LY2456302 and the unlabeled LY2459989. Regional binding potential values derived from the multilinear analysis-1 (MA1) method, as a measure of in vivo specific binding signal, were 2.18, 1.39, 1.08, 1.04, 1.03, 0.59, 0.51, and 0.50, respectively, for the globus pallidus, cingulate cortex, insula, caudate, putamen, frontal cortex, temporal cortex, and thalamus. CONCLUSION The novel PET radioligand (11)C-LY2459989 displayed favorable pharmacokinetic properties, a specific and KOR-selective binding profile, and high specific binding signals in vivo, thus making it a promising PET imaging agent for KOR.
Collapse
Affiliation(s)
- Ming-Qiang Zheng
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Su Jin Kim
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Daniel Holden
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Shu-fei Lin
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Anne Need
- Eli Lilly & Company, Indianapolis, Indiana
| | - Karen Rash
- Eli Lilly & Company, Indianapolis, Indiana
| | | | | | | | - Michael Kapinos
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Kathleen Maloney
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Jim Ropchan
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Richard E Carson
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| | - Yiyun Huang
- PET Center, Department of Diagnostic Radiology, Yale University, New Haven, Connecticut; and
| |
Collapse
|
42
|
Naganawa M, Jacobsen LK, Zheng MQ, Lin SF, Banerjee A, Byon W, Weinzimmer D, Tomasi G, Nabulsi N, Grimwood S, Badura LL, Carson RE, McCarthy TJ, Huang Y. Evaluation of the agonist PET radioligand [¹¹C]GR103545 to image kappa opioid receptor in humans: kinetic model selection, test-retest reproducibility and receptor occupancy by the antagonist PF-04455242. Neuroimage 2014; 99:69-79. [PMID: 24844744 DOI: 10.1016/j.neuroimage.2014.05.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/06/2014] [Accepted: 05/13/2014] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Kappa opioid receptors (KOR) are implicated in several brain disorders. In this report, a first-in-human positron emission tomography (PET) study was conducted with the potent and selective KOR agonist tracer, [(11)C]GR103545, to determine an appropriate kinetic model for analysis of PET imaging data and assess the test-retest reproducibility of model-derived binding parameters. The non-displaceable distribution volume (V(ND)) was estimated from a blocking study with naltrexone. In addition, KOR occupancy of PF-04455242, a selective KOR antagonist that is active in preclinical models of depression, was also investigated. METHODS For determination of a kinetic model and evaluation of test-retest reproducibility, 11 subjects were scanned twice with [(11)C]GR103545. Seven subjects were scanned before and 75 min after oral administration of naltrexone (150 mg). For the KOR occupancy study, six subjects were scanned at baseline and 1.5 h and 8 h after an oral dose of PF-04455242 (15 mg, n=1 and 30 mg, n=5). Metabolite-corrected arterial input functions were measured and all scans were 150 min in duration. Regional time-activity curves (TACs) were analyzed with 1- and 2-tissue compartment models (1TC and 2TC) and the multilinear analysis (MA1) method to derive regional volume of distribution (V(T)). Relative test-retest variability (TRV), absolute test-retest variability (aTRV) and intra-class coefficient (ICC) were calculated to assess test-retest reproducibility of regional VT. Occupancy plots were computed for blocking studies to estimate occupancy and V(ND). The half maximal inhibitory concentration (IC50) of PF-04455242 was determined from occupancies and drug concentrations in plasma. [(11)C]GR103545 in vivo K(D) was also estimated. RESULTS Regional TACs were well described by the 2TC model and MA1. However, 2TC VT was sometimes estimated with high standard error. Thus MA1 was the model of choice. Test-retest variability was ~15%, depending on the outcome measure. The blocking studies with naltrexone and PF-04455242 showed that V(T) was reduced in all regions; thus no suitable reference region is available for the radiotracer. V(ND) was estimated reliably from the occupancy plot of naltrexone blocking (V(ND)=3.4±0.9 mL/cm(3)). The IC50 of PF-04455242 was calculated as 55 ng/mL. [(11)C]GR103545 in vivo K(D) value was estimated as 0.069 nmol/L. CONCLUSIONS [(11)C]GR103545 PET can be used to image and quantify KOR in humans, although it has slow kinetics and variability of model-derived kinetic parameters is higher than desirable. This tracer should be suitable for use in receptor occupancy studies, particularly those that target high occupancy.
Collapse
Affiliation(s)
- Mika Naganawa
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA.
| | | | - Ming-Qiang Zheng
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Shu-Fei Lin
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - David Weinzimmer
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Giampaolo Tomasi
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Nabeel Nabulsi
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Richard E Carson
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Yiyun Huang
- PET Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
43
|
Talwar P, Silla Y, Grover S, Gupta M, Agarwal R, Kushwaha S, Kukreti R. Genomic convergence and network analysis approach to identify candidate genes in Alzheimer's disease. BMC Genomics 2014; 15:199. [PMID: 24628925 PMCID: PMC4028079 DOI: 10.1186/1471-2164-15-199] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 02/21/2014] [Indexed: 01/28/2023] Open
Abstract
Background Alzheimer’s disease (AD) is one of the leading genetically complex and heterogeneous disorder that is influenced by both genetic and environmental factors. The underlying risk factors remain largely unclear for this heterogeneous disorder. In recent years, high throughput methodologies, such as genome-wide linkage analysis (GWL), genome-wide association (GWA) studies, and genome-wide expression profiling (GWE), have led to the identification of several candidate genes associated with AD. However, due to lack of consistency within their findings, an integrative approach is warranted. Here, we have designed a rank based gene prioritization approach involving convergent analysis of multi-dimensional data and protein-protein interaction (PPI) network modelling. Results Our approach employs integration of three different AD datasets- GWL,GWA and GWE to identify overlapping candidate genes ranked using a novel cumulative rank score (SR) based method followed by prioritization using clusters derived from PPI network. SR for each gene is calculated by addition of rank assigned to individual gene based on either p value or score in three datasets. This analysis yielded 108 plausible AD genes. Network modelling by creating PPI using proteins encoded by these genes and their direct interactors resulted in a layered network of 640 proteins. Clustering of these proteins further helped us in identifying 6 significant clusters with 7 proteins (EGFR, ACTB, CDC2, IRAK1, APOE, ABCA1 and AMPH) forming the central hub nodes. Functional annotation of 108 genes revealed their role in several biological activities such as neurogenesis, regulation of MAP kinase activity, response to calcium ion, endocytosis paralleling the AD specific attributes. Finally, 3 potential biochemical biomarkers were found from the overlap of 108 AD proteins with proteins from CSF and plasma proteome. EGFR and ACTB were found to be the two most significant AD risk genes. Conclusions With the assumption that common genetic signals obtained from different methodological platforms might serve as robust AD risk markers than candidates identified using single dimension approach, here we demonstrated an integrated genomic convergence approach for disease candidate gene prioritization from heterogeneous data sources linked to AD. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-199) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Mall Road, Delhi 110 007, India.
| |
Collapse
|
44
|
Yuferov V, Butelman ER, Ho A, Morgello S, Kreek MJ. Neurocognitive and neuroinflammatory correlates of PDYN and OPRK1 mRNA expression in the anterior cingulate in postmortem brain of HIV-infected subjects. J Neuroinflammation 2014; 11:5. [PMID: 24405578 PMCID: PMC3896676 DOI: 10.1186/1742-2094-11-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/21/2013] [Indexed: 01/22/2023] Open
Abstract
Chronic inflammation may contribute to neuropsychological impairments in individuals with HIV, and modulation of this inflammatory response by opiate receptor ligands is important in light of the prevalence of drug use in HIV populations. Exogenous MOR and KOR agonists have differential effects on central nervous system (CNS) immunity and, while some data suggest KOR agonists are immunosuppressive, the KOR agonist dynorphin has been shown to stimulate human monocyte chemotaxis. In this study, we examined mRNA levels of endogenous opioid receptors OPRK1 and OPRM1, prodynorphin (PDYN), macrophage scavenger receptor CD163, and microglia/macrophage marker CD68 in the caudate and anterior cingulate of postmortem brains from HIV-positive and HIV-negative subjects. Brain tissues of HIV-infected (n = 24) and control subjects (n = 15) were obtained from the Manhattan HIV Brain Bank. Quantification of the gene mRNA was performed using SYBR Green RT-PCR. CD68 and CD163 were increased in HIV-positive (HIV+) compared to HIV-negative (HIV-) individuals in both brain regions. There were higher OPRK1 (P <0.005), and lower PDYN mRNA (P <0.005) levels in the anterior cingulate of HIV+ compared to HIV- subjects. This difference between the clinical groups was not found in the caudate. There was no difference in the levels of OPRM1 mRNA between HIV+ and HIV- subjects. Using linear regression analysis, we examined the relationship of OPRK1 and PDYN mRNA levels in the HIV+ subjects with seven cognitive domain T scores of a neuropsychological test battery. Within the HIV+ subjects, there was a positive correlation between anterior cingulate PDYN mRNA levels and better T-scores in the motor domain. Within the HIV+ subjects there were also positive correlations of both OPRK1 and PDYN mRNA levels with the anti-inflammatory marker CD163, but not with proinflammatory CD68 levels. In this setting, decreased PDYN mRNA may reflect a homeostatic mechanism to reduce monocyte migration, accompanied by compensatory increases in the cognate receptor (KOR) to dampen pro-inflammatory responses. It is possible that enhanced neuroprotection and better motor performance are associated with higher levels of dynorphin and the recruitment of neuroprotective CD163-positive macrophages. Further studies are needed to test this hypothesis.
Collapse
Affiliation(s)
- Vadim Yuferov
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | | | | | | | | |
Collapse
|
45
|
Arneric SP, Laird JM, Chappell AS, Kennedy JD. Tailoring chronic pain treatments for the elderly: are we prepared for the challenge? Drug Discov Today 2014; 19:8-17. [DOI: 10.1016/j.drudis.2013.08.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/08/2013] [Accepted: 08/22/2013] [Indexed: 12/21/2022]
|
46
|
Loriga G, Lazzari P, Ruiu S, Marchese G, Manca I, Casu GL, Dessì C, Pinna GA, Asproni B, Murineddu G. Synthesis and biological evaluation of novel delta (δ) opioid receptor ligands with diazatricyclodecane skeletons. Eur J Med Chem 2013; 69:413-26. [PMID: 24090913 DOI: 10.1016/j.ejmech.2013.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 12/01/2022]
Abstract
Considering the interesting pharmacological profile of the delta (δ) selective opioid agonist compound SNC-80, conformationally constrained analogs containing two diazatricyclodecane ring systems in place of dimethylpiperazine core motif were synthesized. The compounds showed subnanomolar or low nanomolar δ opioid receptor binding affinity. Depending upon the substituents on the diazatricyclodecane ring, these compounds displayed varying selectivity for δ opioid receptor over μ and κ receptors. Amongst the novel compounds, 1Aa showed the more interesting biological profile, with higher δ affinity and selectivity compared to SNC-80. The δ receptor agonist profile and antinociceptive activity of 1Aa were confirmed using ex-vivo (isolated mouse vas deferens) and in vivo (tail flick) assays.
Collapse
Affiliation(s)
- Giovanni Loriga
- C.N.R. Istituto di Farmacologia Traslazionale, UOS Cagliari, Edificio 5, Loc. Piscinamanna, 09010 Pula, CA, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Garland EL, Froeliger B, Zeidan F, Partin K, Howard MO. The downward spiral of chronic pain, prescription opioid misuse, and addiction: cognitive, affective, and neuropsychopharmacologic pathways. Neurosci Biobehav Rev 2013; 37:2597-607. [PMID: 23988582 DOI: 10.1016/j.neubiorev.2013.08.006] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/15/2013] [Indexed: 12/19/2022]
Abstract
Prescription opioid misuse and addiction among chronic pain patients are emerging public health concerns of considerable significance. Estimates suggest that more than 10% of chronic pain patients misuse opioid analgesics, and the number of fatalities related to nonmedical or inappropriate use of prescription opioids is climbing. Because the prevalence and adverse consequences of this threat are increasing, there is a pressing need for research that identifies the biobehavioral risk chain linking chronic pain, opioid analgesia, and addictive behaviors. To that end, the current manuscript draws upon current neuropsychopharmacologic research to provide a conceptual framework of the downward spiral leading to prescription opioid misuse and addiction among chronic pain patients receiving opioid analgesic pharmacotherapy. Addictive use of opioids is described as the outcome of a cycle initiated by chronic pain and negative affect and reinforced by opioidergic-dopamingeric interactions, leading to attentional hypervigilance for pain and drug cues, dysfunctional connectivity between self-referential and cognitive control networks in the brain, and allostatic dysregulation of stress and reward circuitry. Implications for clinical practice are discussed; multimodal, mindfulness-oriented treatment is introduced as a potentially effective approach to disrupting the downward spiral and facilitating recovery from chronic pain and opioid addiction.
Collapse
Affiliation(s)
- Eric L Garland
- Supportive Oncology & Survivorship Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States; College of Social Work, University of Utah, Salt Lake City, UT, United States.
| | | | | | | | | |
Collapse
|
48
|
Ménard C, Herzog H, Schwarzer C, Quirion R. Possible role of dynorphins in Alzheimer's disease and age-related cognitive deficits. NEURODEGENER DIS 2013; 13:82-5. [PMID: 23970097 DOI: 10.1159/000353848] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 06/18/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Expression of dynorphin, an endogenous opioid peptide, increases with age and has been associated with cognitive deficits in rodents. Elevated dynorphin levels have been reported in postmortem samples from Alzheimer's disease (AD) patients, and prodynorphin (PDYN) gene polymorphisms might be linked to cognitive function in the elderly. Activation of κ-opioid receptors by dynorphins has been associated with stress-related memory impairments. Interestingly, these peptides can also modulate glutamate neurotransmission and may affect synaptic plasticity underlying memory formation. N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazol-propionate (AMPA) ionotropic glutamate receptor levels generally decrease with aging, and their function is impaired in AD. METHODS Here, we compared the impact of aging on ionotropic glutamate receptor levels in the hippocampal formation of wild-type (WT) and Pdyn knock-out (KO) mice. RESULTS We observed a significant reduction in GluR1 and GluR2 AMPA receptor subunits in the hippocampal formation of 18- to 25-month-old WT mice in comparison with 6-month-old mice. Conversely, the GluR1 protein level was maintained in old Pdyn KO mice, and the NMDA NR2B subunit level was increased by 42% when compared to old WT animals. CONCLUSIONS These results suggest that elevated dynorphin expression occurring during aging and AD may mediate cognitive deficits by altering the glutamatergic system integrity.
Collapse
Affiliation(s)
- Caroline Ménard
- Douglas Mental Health University Institute, McGill University, Montreal, Qué., Canada
| | | | | | | |
Collapse
|
49
|
Pharmacological traits of delta opioid receptors: pitfalls or opportunities? Psychopharmacology (Berl) 2013; 228:1-18. [PMID: 23649885 PMCID: PMC3679311 DOI: 10.1007/s00213-013-3129-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/15/2013] [Indexed: 12/11/2022]
Abstract
RATIONALE Delta opioid receptors (DORs) have been considered as a potential target to relieve pain as well as treat depression and anxiety disorders and are known to modulate other physiological responses, including ethanol and food consumption. A small number of DOR-selective drugs are in clinical trials, but no DOR-selective drugs have been approved by the Federal Drug Administration and some candidates have failed in phase II clinical trials, highlighting current difficulties producing effective delta opioid-based therapies. Recent studies have provided new insights into the pharmacology of the DOR, which is often complex and at times paradoxical. OBJECTIVE This review will discuss the existing literature focusing on four aspects: (1) Two DOR subtypes have been postulated based on differences in pharmacological effects of existing DOR-selective ligands. (2) DORs are expressed ubiquitously throughout the body and central nervous system and are, thus, positioned to play a role in a multitude of diseases. (3) DOR expression is often dynamic, with many reports of increased expression during exposure to chronic stimuli, such as stress, inflammation, neuropathy, morphine, or changes in endogenous opioid tone. (4) A large structural variety in DOR ligands implies potential different mechanisms of activating the receptor. CONCLUSION The reviewed features of DOR pharmacology illustrate the potential benefit of designing tailored or biased DOR ligands.
Collapse
|
50
|
Bailey CR, Cordell E, Sobin SM, Neumeister A. Recent progress in understanding the pathophysiology of post-traumatic stress disorder: implications for targeted pharmacological treatment. CNS Drugs 2013; 27:221-32. [PMID: 23483368 PMCID: PMC3629370 DOI: 10.1007/s40263-013-0051-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a common and chronic anxiety disorder that can result after exposure to a traumatic event. Though our understanding of the aetiology of PTSD is incomplete, several neurobiological systems have been implicated in the pathophysiology and vulnerability towards developing PTSD after trauma exposure. We aimed to provide a concise review of benchmark findings in important neurobiological systems related to the aetiology and maintenance of PTSD symptomology. Specifically, we discuss functional aetiologies in the noradrenergic, serotonergic, endogenous cannabinoid and opioid systems as well as the hypothalamic-pituitary adrenal (HPA) axis. This article provides a succinct framework to appreciate the current understanding of neurobiological mechanisms related to the pathophysiology of PTSD and how these findings may impact the development of future, targeted pharmacological treatments for this debilitating disorder.
Collapse
|