1
|
Gugula A, Sambak P, Trenk A, Drabik S, Nogaj A, Soltys Z, Gundlach AL, Blasiak A. Early-life adversity alters adult nucleus incertus neurons: implications for neuronal mechanisms of increased stress and compulsive behavior vulnerability. Neuropsychopharmacology 2025:10.1038/s41386-025-02089-0. [PMID: 40114019 DOI: 10.1038/s41386-025-02089-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
Early-life stress (ELS) arising from physical and emotional abuse disrupts normal brain development and impairs hypothalamic-pituitary-adrenal axis function, increasing the risk of psychopathological disorders and compulsive behaviors in adulthood. However, the underlying neural mechanisms remain unclear. The brainstem nucleus incertus (NI) is a highly stress-sensitive locus, involved in behavioral activation and stress-induced reward (food/alcohol) seeking, but its sensitivity to ELS remains unexplored. We used neonatal maternal separation stress in rats as a model for ELS and examined its impact on stress-related mRNA and neuropeptide expression in the NI, using fluorescent in situ hybridization and immunohistochemistry, respectively. Using whole-cell, patch-clamp recordings we determined the influence of ELS on the synaptic activity, excitability, and electrophysiological properties of NI neurons. Using c-Fos protein expression we also assessed the impact of ELS on the sensitivity of NI neurons to acute restraint stress in adulthood. ELS weakened the acute stress responsiveness of NI neurons, and caused dendritic shrinkage, impaired synaptic transmission and altered electrophysiological properties of NI neurons in a cell-type-specific manner. Additionally, ELS increased the expression of mRNA encoding corticotropin-releasing hormone receptor type 1 and the nerve-growth factor receptor, TrkA in adult NI. The multiple, cell-type specific changes in the expression of neuropeptides and molecules associated with stress and substance abuse in the NI, as well as impairments in NI neuron morphology and electrophysiology caused by ELS and observed in the adult brain, may contribute to the increased susceptibility to stress and compulsive behaviors observed in individuals with a history of ELS.
Collapse
Affiliation(s)
- Anna Gugula
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Patryk Sambak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Aleksandra Trenk
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Sylwia Drabik
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Aleksandra Nogaj
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Zbigniew Soltys
- Laboratory of Experimental Neuropathology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, and Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Anna Blasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
2
|
Kuo SC, Lin CL, Tsou CC, Yeh YW, Yang BZ, Chen CY, Huang CY, Huang SY. Nerve growth factor gene polymorphisms may be associated with heroin dependence in women but do not mediate specific personality traits. Eur Arch Psychiatry Clin Neurosci 2025; 275:487-498. [PMID: 39311999 PMCID: PMC11976897 DOI: 10.1007/s00406-024-01906-9] [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: 03/09/2024] [Accepted: 09/07/2024] [Indexed: 10/26/2024]
Abstract
Heroin dependence (HD) is a complex disease with a substantial genetic contribution and is associated with traits of impulsivity and specific personality traits. The neurotrophic factor nerve growth factor (NGF) may mediate the reward processes in HD. This study aims to investigate whether NGF gene polymorphisms are associated with the co-occurrence of HD and impulsivity/specific personality traits in HD patients. To minimize the potential confounding effects of population stratification, we selected a homogeneous Han Chinese population and recruited 1364 participants (831 HD patients and 533 healthy controls). In addition, 163 female HD patients completed the Chinese version of the Barratt Impulsiveness Scale Version 11 (BIS-11), and 440 HD patients completed the Chinese version of the Tridimensional Personality Questionnaire (TPQ) for subsequent analysis. We identified three polymorphisms with altered allele and genotype frequency in HD patients versus controls (p = 0.035 for rs2254527; p = 0.005 for rs6678788; p = 0.006 for rs7523654), especially in the female subgroup. Four associations identified via haplotype analysis were significant in the female subgroup (p = 0.003 for T-T-A haplotype and p = 0.002 for C-C-A haplotype in block 1; p = 0.011 for T-T haplotype and p = 0.009 for C-T haplotypes in block 2), but not in the male subgroup. Male HD patients had higher novelty-seeking (NS) scores, and female HD patients had higher harm avoidance (HA) scores. However, there was no significant association between the selected NGF polymorphisms and BIS or TPQ scores in HD patients. NGF variants may contribute to the risk of HD development in females but do not mediate the relationship between impulsivity and specific personality traits in the female population.
Collapse
Affiliation(s)
- Shin-Chang Kuo
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Long Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Psychiatry, Taoyuan Armed Forces General Hospital, Hsinchu Branch, Hsinchu, Taiwan
| | - Chang-Chih Tsou
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Wei Yeh
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Bao-Zhu Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, and VA CT Healthcare Center, West Haven, CT, USA
| | - Chun-Yen Chen
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yun Huang
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - San-Yuan Huang
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
3
|
Jokara Z, Khatamsaz S, Alaei H, Shariati M. Effect of electrical stimulation of central nucleus of the amygdala on morphine conditioned place preference in male rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:604-610. [PMID: 35911639 PMCID: PMC9282746 DOI: 10.22038/ijbms.2022.62133.13751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/09/2022] [Indexed: 11/28/2022]
Abstract
Objectives The central nucleus of the amygdala (CeA) is one of the most important areas for the morphine reward system. This study investigated the effect of electrical stimulation of CeA on morphine conditioned place preference (CPP) in male rats. Materials and Methods After anesthetizing male Wistar rats, both electrode and cannula were implanted into CeA for stimulating (low intensity: 25 μA, and high intensity: 150 μA) and injecting (lidocaine and dopamine D2 receptor antagonist), respectively. Then, CPP induced by effective (5 mg/kg) and ineffective (0.5 mg/kg) doses of morphine was evaluated for five consecutive days (n = 6 / group). Results The low electrical stimulation intensity of 25 μA suppressed both acquisition and expression phases, but the high intensity of 150 µA attenuated only the expression phase. On the other hand, intra-CeA administration of dopamine D2 receptor antagonist, eticlopride (2 µg/rat), with the effective dose of morphine, decreased CPP. In addition, infusion of lidocaine into the CeA inhibited morphine-induced CPP in both acquisition and expression phases with the effective dose of morphine. Conclusion Electrical stimulation of the CeA may play an important role in attenuating morphine induced CPP via possible changes in neurotransmitters involved in the reward system such as dopamine (DA) and gamma-aminobutyric acid (GABA).
Collapse
Affiliation(s)
- Zahra Jokara
- Department of Biology, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Saeed Khatamsaz
- Department of Biology, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - HojjatAllah Alaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrdad Shariati
- Department of Biology, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| |
Collapse
|
4
|
Zhang H, Qu M, Gorur A, Sun Z, Cata JP, Chen W, Miao C. Association of Mu-Opioid Receptor(MOR) Expression and Opioids Requirement With Survival in Patients With Stage I-III Pancreatic Ductal Adenocarcinoma. Front Oncol 2021; 11:686877. [PMID: 34222012 PMCID: PMC8249918 DOI: 10.3389/fonc.2021.686877] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/07/2021] [Indexed: 01/16/2023] Open
Abstract
Background The use of opioids in patients with metastatic pancreatic ductal adenocarcinoma (PDAC) is associated with shorter survival and not dependent on the expression of the mu-opioid receptor (MOR). The role of opioid use and MOR expression in stage I-III PDAC has not been investigated. Methods We conducted retrospective study in patients with stage I-III PDAC. MOR expression and OPRM1 gene expression in tumour tissue and non-tumour tissue was measured. Primary endpoints were overall survival (OS) and disease-free survival (DFS). Secondary endpoints included perineural invasion, intraoperative sufentanil consumption, and length of stay. We performed a subgroup group analysis to evaluate the interaction between levels of MOR expression, amount of opioids use (high versus low) and its association with survival. Results A total of 236 patients were enrolled in this study.There were no significantly difference in OS rates in patients with high versus low levels of MOR (1-year OS: 65.2% versus 70.6%, P=0.064; 3-year: 31.4% versus 35.8%, P=0.071; 5-year: 19.4% versus. 16.2%, P=0.153, respectively) in the tumours. The DFS rates between the groups were no significantly difference. Of note, a high expression of MOR combined with high opioid consumption was associated with poor prognosis in stage I-III PDAC patients. Tumor expressing high levels of MOR show higher rates of perineural invasion. Conclusion MOR is not an independent predictor of poor survival in stage I-III PDAC but associated with perineural invasion. Patients requiring high amounts of opioids intraoperatively show worse outcome if they are expressing high levels of MOR.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Anaesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengdi Qu
- Department of Anaesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Aysegul Gorur
- Department of Anaesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Centre, Houston, TX, United States.,Anaesthesiology and Surgical Oncology Research Group, Houston, TX, United States
| | - Zhirong Sun
- Department of Anaesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Juan P Cata
- Department of Anaesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Centre, Houston, TX, United States.,Anaesthesiology and Surgical Oncology Research Group, Houston, TX, United States
| | - Wankun Chen
- Department of Anaesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Fudan Zhangjiang Institute, Shanghai, China
| | - Changhong Miao
- Department of Anaesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Neurotrophin signalling in amygdala-dependent cued fear learning. Cell Tissue Res 2020; 382:161-172. [PMID: 32845430 PMCID: PMC7529623 DOI: 10.1007/s00441-020-03260-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022]
Abstract
The amygdala is a central hub for fear learning assessed by Pavlovian fear conditioning. Indeed, the prevailing hypothesis that learning and memory are mediated by changes in synaptic strength was shown most convincingly at thalamic and cortical afferents to the lateral amygdala. The neurotrophin brain-derived neurotrophic factor (BDNF) is known to regulate synaptic plasticity and memory formation in many areas of the mammalian brain including the amygdala, where BDNF signalling via tropomyosin-related kinase B (TrkB) receptors is prominently involved in fear learning. This review updates the current understanding of BDNF/TrkB signalling in the amygdala related to fear learning and extinction. In addition, actions of proBDNF/p75NTR and NGF/TrkA as well as NT-3/TrkC signalling in the amygdala are introduced.
Collapse
|
6
|
Epigenetic upregulation of hippocampal CXCL12 contributes to context spatial memory-associated morphine conditioning. Brain Behav Immun 2020; 84:72-79. [PMID: 31751616 DOI: 10.1016/j.bbi.2019.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/15/2019] [Accepted: 11/17/2019] [Indexed: 11/21/2022] Open
Abstract
Conditioned place preference (CPP) is a learned behavior, in which animals learn to associate environmental contexts with rewarding effects. The formation of CPP is an integrated outcome of multiple learning processes. Although multiple anatomical substrates underlying this contextual learning have been proposed, it remains unknown whether a specific molecular signaling pathway within CA1 mediates context learning associated with morphine conditioning. Here, we showed that repeated context learning associated with morphine conditioning significantly increased CXCL12 levels in hippocampal CA1 neurons, and the inhibition of CXCL12 expression ameliorated the CPP behavior following context exposure with morphine conditioning. Additionally, repeated context exposure with morphine conditioning increased the phosphorylation of STAT3 and the acetylation of histone H4 in CXCL12-expressing neurons in CA1. Immunoprecipitation and chromatin immunoprecipitation assays demonstrated that repeated context exposure with morphine conditioning increased the binding of STAT3 to the CXCL12 gene promoter and the interaction between STAT3 and p300, which contributed to the enhanced transcription of CXCL12 by increasing the acetylation of histone H4 in the CXCL12 gene promoter. The inhibition of STAT3 by intrathecal injection of S3I-201 suppressed the acetylation of histone H4. These data demonstrated the epigenetic upregulation of CXCL12 following repeated context exposure with morphine conditioning, which potentially contributed to the spatial memory consolidation associated with conditioned place preference induced by morphine conditioning.
Collapse
|
7
|
Kumar A, Kumar P, Pareek V, Faiq MA, Narayan RK, Raza K, Prasoon P, Sharma VK. Neurotrophin mediated HPA axis dysregulation in stress induced genesis of psychiatric disorders: Orchestration by epigenetic modifications. J Chem Neuroanat 2019; 102:101688. [PMID: 31568825 DOI: 10.1016/j.jchemneu.2019.101688] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/22/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Abstract
Apart from their established role in embryonic development, neurotrophins (NTs) have diverse functions in the nervous system. Their role in the integration of physiological and biochemical aspects of the nervous system is currently attracting much attention. Based on a systematic analysis of the literature, we here propose a new paradigm that, by exploiting a novel role of NTs, may help explain the genesis of stress-related psychiatric disorders, opening new avenues for better management of the same. We hypothesize that NTs as an integrated network play a crucial role in maintaining an indivdual's psychological wellbeing. Given the evidence that stress can induce chronic disruption of the hypothalamic-pituitary-adrenal (HPA) axis which, in turn, is causally linked to several psychiatric disorders, this function may be mediated through the homeostatic mechanisms governing regulation of this axis. In fact, NTs, such as nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) are known to participate in neuroendocrine regulation. Recent studies suggest epigenetic modification of NT-HPA axis interplay in the precipitation of psychiatric disorders. Our article highlights why this new knowledge regarding NTs should be considered in the etiogenesis and treatment of stress-induced psychopathology.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India; Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India; Etiologically Elusive Disorders Research Network (EEDRN), India.
| | - Pavan Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India; Developmental Neurogenetics Lab, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Vikas Pareek
- Computational Neuroscience and Neuroimaging Division, National Brain Research Centre (NBRC), Manesar, Haryana, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Muneeb A Faiq
- Neuroimaging and Visual Science Laboratory, New York University (NYU) Langone Health, NYU School of Medicine, New York, NY 10016, USA; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Ravi K Narayan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Khursheed Raza
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Pranav Prasoon
- Pittsburgh Centre for Pain Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Etiologically Elusive Disorders Research Network (EEDRN), India
| | - Vivek K Sharma
- Department of Physiology, Government Institute of Medical Sciences (GIMS), Greater Noida, Kasna, Uttar Pradesh, India; Etiologically Elusive Disorders Research Network (EEDRN), India
| |
Collapse
|
8
|
Martínez-Rivera FJ, Martínez NA, Martínez M, Ayala-Pagán RN, Silva WI, Barreto-Estrada JL. Neuroplasticity transcript profile of the ventral striatum in the extinction of opioid-induced conditioned place preference. Neurobiol Learn Mem 2019; 163:107031. [PMID: 31173919 PMCID: PMC6689252 DOI: 10.1016/j.nlm.2019.107031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/27/2019] [Accepted: 06/03/2019] [Indexed: 01/03/2023]
Abstract
Persistent drug-seeking behavior has been associated with deficits in neural circuits that regulate the extinction of addictive behaviors. Although there is extensive data that associates addiction phases with neuroplasticity changes in the reward circuit, little is known about the underlying mechanisms of extinction learning of opioid associated cues. Here, we combined morphine-conditioned place preference (CPP) with real-time polymerase chain reaction (RT-PCR) to identify the effects of extinction training on the expression of genes (mRNAs) associated with synaptic plasticity and opioid receptors in the ventral striatum/nucleus accumbens (VS/NAc). Following morphine extinction training, we identified two animal subgroups showing either extinction (low CPP) or extinction-resistance (high CPP). A third group were conditioned to morphine but did not receive extinction training (sham-extinction; high CPP). RT-PCR results showed that brain derived neurotrophic factor (Bdnf) was upregulated in rats showing successful extinction. Conversely, the lack of extinction training (sham-extinction) upregulated genes associated with kinases (Camk2g), neurotrophins (Ngfr), synaptic connectivity factors (Ephb2), glutamate neurotransmission (Grm8) and opioid receptors (μ1, Δ1). To further identify genes modulated by morphine itself, comparisons with their saline-counterparts were performed. Results revealed that Bdnf was consistently upregulated in the extinction group. Alternatively, widespread gene modulation was observed in the group with lack of extinction training (i.e. Drd2, Cnr1, Creb, μ1, Δ1) and the group showing extinction resistance (i.e. Crem, Rheb, Tnfa). Together, our study builds on the identification of putative genetic markers for the extinction learning of drug-associated cues.
Collapse
Affiliation(s)
- Freddyson J Martínez-Rivera
- Department of Anatomy and Neurobiology, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00936, USA
| | - Namyr A Martínez
- Department of Physiology and Biophysics, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00936, USA; Molecular Sciences Building, University of Puerto Rico, San Juan, PR 00926, USA
| | - Magdiel Martínez
- Department of Physiology and Biophysics, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00936, USA; Molecular Sciences Building, University of Puerto Rico, San Juan, PR 00926, USA
| | - Roxsana N Ayala-Pagán
- Department of Anatomy and Neurobiology, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00936, USA
| | - Walter I Silva
- Department of Physiology and Biophysics, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00936, USA; Molecular Sciences Building, University of Puerto Rico, San Juan, PR 00926, USA
| | - Jennifer L Barreto-Estrada
- Department of Anatomy and Neurobiology, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00936, USA.
| |
Collapse
|
9
|
Rouhani F, Khodarahmi P, Naseh V. NGF, BDNF and Arc mRNA Expression in the Hippocampus of Rats After Administration of Morphine. Neurochem Res 2019; 44:2139-2146. [DOI: 10.1007/s11064-019-02851-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/21/2022]
|
10
|
Wu J, Hocevar M, Bie B, Foss JF, Naguib M. Cannabinoid Type 2 Receptor System Modulates Paclitaxel-Induced Microglial Dysregulation and Central Sensitization in Rats. THE JOURNAL OF PAIN 2019; 20:501-514. [DOI: 10.1016/j.jpain.2018.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/12/2018] [Accepted: 10/24/2018] [Indexed: 12/30/2022]
|
11
|
Marton S, González B, Rodríguez-Bottero S, Miquel E, Martínez-Palma L, Pazos M, Prieto JP, Rodríguez P, Sames D, Seoane G, Scorza C, Cassina P, Carrera I. Ibogaine Administration Modifies GDNF and BDNF Expression in Brain Regions Involved in Mesocorticolimbic and Nigral Dopaminergic Circuits. Front Pharmacol 2019; 10:193. [PMID: 30890941 PMCID: PMC6411846 DOI: 10.3389/fphar.2019.00193] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/14/2019] [Indexed: 01/07/2023] Open
Abstract
Ibogaine is an atypical psychedelic alkaloid, which has been subject of research due to its reported ability to attenuate drug-seeking behavior. Recent work has suggested that ibogaine effects on alcohol self-administration in rats are related to the release of Glial cell Derived Neurotrophic Factor (GDNF) in the Ventral Tegmental Area (VTA), a mesencephalic region which hosts the soma of dopaminergic neurons. Although previous reports have shown ibogaine’s ability to induce GDNF expression in rat midbrain, there are no studies addressing its effect on the expression of GDNF and other neurotrophic factors (NFs) such as Brain Derived Neurotrophic Factor (BDNF) or Nerve Growth Factor (NGF) in distinct brain regions containing dopaminergic neurons. In this work, we examined the effect of ibogaine acute administration on the expression of these NFs in the VTA, Prefrontal Cortex (PFC), Nucleus Accumbens (NAcc) and the Substantia Nigra (SN). Rats were i.p. treated with ibogaine 20 mg/kg (I20), 40 mg/kg (I40) or vehicle, and NFs expression was analyzed after 3 and 24 h. At 24 h an increase of the expression of the NFs transcripts was observed in a site and dose dependent manner. Only for I40, GDNF was selectively upregulated in the VTA and SN. Both doses elicited a large increase in the expression of BDNF transcripts in the NAcc, SN and PFC, while in the VTA a significant effect was found only for I40. Finally, NGF mRNA was upregulated in all regions after I40, while I20 showed a selective upregulation in PFC and VTA. Regarding protein levels, an increase of GDNF was observed in the VTA only for I40 but no significant increase for BDNF was found in all the studied areas. Interestingly, an increase of proBDNF was detected in the NAcc for both doses. These results show for the first time a selective increase of GDNF specifically in the VTA for I40 but not for I20 after 24 h of administration, which agrees with the effective dose found in previous self-administration studies in rodents. Further research is needed to understand the contribution of these changes to ibogaine’s ability to attenuate drug-seeking behavior.
Collapse
Affiliation(s)
- Soledad Marton
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Bruno González
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Sebastián Rodríguez-Bottero
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Ernesto Miquel
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Laura Martínez-Palma
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mariana Pazos
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - José Pedro Prieto
- Departamento de Neurofarmacología Experimental, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Paola Rodríguez
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY, United States
| | - Gustavo Seoane
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Scorza
- Departamento de Neurofarmacología Experimental, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Patricia Cassina
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Ignacio Carrera
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
12
|
Tsou CC, Kuo SC, Chen CY, Lu RB, Wang TJ, Huang SY. NGF gene polymorphisms are not associated with heroin dependence in a Taiwanese male population. Am J Addict 2018; 27:516-523. [PMID: 30070410 DOI: 10.1111/ajad.12785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 07/19/2018] [Accepted: 07/22/2018] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Heroin dependence (HD) is a chronic relapsing brain illness with substantial heritability. Nerve growth factor (NGF) is a crucial modulator in the neurodevelopment, and may be a key mediator of reward processes in HD. The purpose of this genetic study was to investigate whether NGF gene polymorphisms associate with the occurrence of HD and the specific personality traits of patients with HD. METHODS We selected a homogeneous Han Chinese male population to overcome possible confounding effects of population and gender. For the study, 272 HD patients and 141 controls completed the Tridimensional Personality Questionnaire to evaluate their personality traits. In addition, a further sample 303 HD patients and 204 controls was added (with totally 920 participants) for the gene association and genotype-phenotype interaction studies. RESULTS Patients with HD had higher novelty seeking (NS) and harm avoidance (HA) scores than healthy subjects. Nonetheless, NGF gene polymorphisms did not associate with specific personality traits in HD patients and controls. There is no significant difference in NGF gene polymorphisms between patients with HD and controls. DISCUSSION AND CONCLUSIONS The NGF gene may neither contribute to the risk of development of HD, nor mediate the relationship between specific personality traits and HD in Han Chinese male population. SCIENTIFIC SIGNIFICANCE Patients with HD had higher novelty seeking (NS) and harm avoidance (HA) scores than healthy subjects. However, none of the polymorphisms in the NGF gene affected the NS and HA scores in both patients and healthy subjects. (Am J Addict 2018;27:516-523).
Collapse
Affiliation(s)
- Chang-Chih Tsou
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Shin-Chang Kuo
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Chun-Yen Chen
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Ru-Band Lu
- Department of Psychiatry, College of Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Tso-Jen Wang
- Department of Psychiatry, Jianan Psychiatric Center, Ministry of Health and Welfare, Tainan, Taiwan, R.O.C
| | - San-Yuan Huang
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
| |
Collapse
|
13
|
Effects of docosahexaenoic acid on locomotor activity in ethanol-treated HIV-1 transgenic rats. J Neurovirol 2017; 24:88-97. [PMID: 29260441 DOI: 10.1007/s13365-017-0597-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 10/26/2017] [Accepted: 11/01/2017] [Indexed: 01/17/2023]
Abstract
Binge drinking affects the onset and progression of human immunodeficiency virus (HIV)-associated neurological disorders. The HIV-1 transgenic (HIV-1Tg) rat was created with a gag- and pol-deleted HIV-1 viral genome to mimic HIV-infected patients receiving combination anti-retroviral therapy (cART). Docosahexaenoic acid (DHA) is a marine compound that modulates inflammatory responses. Using HIV-1Tg rats subjected to binge exposure to ethanol (EtOH), this study examined whether DHA could reduce the detrimental neurological effects of EtOH and HIV proteins. Young adult male HIV-1Tg and F344 control rats received 4 mL/kg/day saline as a control (Saline group), 20 mg/kg/day DHA (DHA group), 4.8 g/kg/day 52% w/v EtOH (EtOH group), or 4.8 g/kg/day 52% w/v EtOH and 20 mg/kg/d DHA (DHA + EtOH group) by gavage for 5 weeks (n = 6 per group). EtOH was administrated on days 5, 6, and 7 of each week. Locomotor activity (LMA) was assessed using open field tests before and 45, 90, 135, and 180 min after each treatment. Repeated binge EtOH exposure gradually decreased LMA measured before daily treatments in HIV-1Tg and F344 rats, an effect that was reversed by DHA only in the HIV-1Tg rats. Decreased LMA of rats after treatment and under the influence of EtOH was less pronounced, and the reversal effect of DHA did not reach statistical significance. The plasma endotoxin level was significantly higher in HIV-1Tg rats than in F344 rats. IL-6 and IL-18 expression in the striatum was significantly higher in the HIV-1Tg EtOH group than in the F344 EtOH group. DHA significantly decreased the high levels of IL-6, IL-18, and NF-κB expression observed in the HIV-1Tg EtOH group. DHA appears to ameliorate inflammation and consequently lessen the reductions in LMA produced by the combination of EtOH and HIV-1 viral proteins.
Collapse
|
14
|
Kumar A, Pareek V, Faiq MA, Kumar P, Raza K, Prasoon P, Dantham S, Mochan S. Regulatory role of NGFs in neurocognitive functions. Rev Neurosci 2017; 28:649-673. [DOI: 10.1515/revneuro-2016-0031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
AbstractNerve growth factors (NGFs), especially the prototype NGF and brain-derived neurotrophic factor (BDNF), have a diverse array of functions in the central nervous system through their peculiar set of receptors and intricate signaling. They are implicated not only in the development of the nervous system but also in regulation of neurocognitive functions like learning, memory, synaptic transmission, and plasticity. Evidence even suggests their role in continued neurogenesis and experience-dependent neural network remodeling in adult brain. They have also been associated extensively with brain disorders characterized by neurocognitive dysfunction. In the present article, we aimed to make an exhaustive review of literature to get a comprehensive view on the role of NGFs in neurocognitive functions in health and disease. Starting with historical perspective, distribution in adult brain, implied molecular mechanisms, and developmental basis, this article further provides a detailed account of NGFs’ role in specified neurocognitive functions. Furthermore, it discusses plausible NGF-based homeostatic and adaptation mechanisms operating in the pathogenesis of neurocognitive disorders and has presents a survey of such disorders. Finally, it elaborates on current evidence and future possibilities in therapeutic applications of NGFs with an emphasis on recent research updates in drug delivery mechanisms. Conclusive remarks of the article make a strong case for plausible role of NGFs in comprehensive regulation of the neurocognitive functions and pathogenesis of related disorders and advocate that future research should be directed to explore use of NGF-based mechanisms in the prevention of implicated diseases as well as to target these molecules pharmacologically.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
- Department of Anatomy, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Karaikal, Puducherry 609602, India
| | - Vikas Pareek
- Computational Neuroscience and Neuroimaging Division, National Brain Research Centre (NBRC), Manesar, Haryana 122051, India
| | - Muneeb A. Faiq
- Department of Ophthalmology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pavan Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Khursheed Raza
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pranav Prasoon
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Subrahamanyam Dantham
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sankat Mochan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| |
Collapse
|
15
|
Norman H, D'Souza MS. Endogenous opioid system: a promising target for future smoking cessation medications. Psychopharmacology (Berl) 2017; 234:1371-1394. [PMID: 28285326 DOI: 10.1007/s00213-017-4582-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 02/24/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Nicotine addiction continues to be a health challenge across the world. Despite several approved medications, smokers continue to relapse. Several human and animal studies have evaluated the role of the endogenous opioid system as a potential target for smoking cessation medications. METHODS In this review, studies that have elucidated the role of the mu (MORs), delta (DORs), and kappa (KORs) opioid receptors in nicotine reward, nicotine withdrawal, and reinstatement of nicotine seeking will be discussed. Additionally, the review will discuss discrepancies in the literature and therapeutic potential of the endogenous opioid system, and suggest studies to address gaps in knowledge with respect to the role of the opioid receptors in nicotine dependence. RESULTS Data available till date suggest that blockade of the MORs and DORs decreased the rewarding effects of nicotine, while activation of the MORs and DORs decreased nicotine withdrawal-induced aversive effects. In contrast, activation of the KORs decreased the rewarding effects of nicotine, while blockade of the KORs decreased nicotine withdrawal-induced aversive effects. Interestingly, blockade of the MORs and KORs attenuated reinstatement of nicotine seeking. In humans, MOR antagonists have shown benefits in select subpopulations of smokers and further investigation is required to realize their full therapeutic potential. CONCLUSION Future work must assess the influence of polymorphisms in opioid receptor-linked genes in nicotine dependence, which will help in both identifying individuals vulnerable to nicotine addiction and the development of opioid-based smoking cessation medications. Overall, the endogenous opioid system continues to be a promising target for future smoking cessation medications.
Collapse
Affiliation(s)
- Haval Norman
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA
| | - Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA.
| |
Collapse
|
16
|
Activation of the Extracellular Signal-Regulated Kinase in the Amygdale Modulates Fentanyl-Induced Hypersensitivity in Rats. THE JOURNAL OF PAIN 2016; 18:188-199. [PMID: 27838497 DOI: 10.1016/j.jpain.2016.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 10/08/2016] [Accepted: 10/24/2016] [Indexed: 11/23/2022]
Abstract
Opioid-induced hyperalgesia (OIH) is one of the major problems associated with use of opioids in perioperative and chronic pain management. The mechanism underlying this paradoxical phenomenon needs to be fully elucidated. Laterocapsular division of the central nucleus of amygdale (CeLC) has emerged as an important brain center for pain modulation, so we hypothesize that the activation of extracellular signal-regulated kinase (ERK) in CeLC may modulate OIH through strengthening synaptic transmission between neurons in the CeLC. Phospho-ERK in CeLC was first found to be increased significantly in OIH rats induced by repeated subcutaneous injection of fentanyl. Blockade of this fentanyl-induced ERK activation by microinjection of U0126, an ERK inhibitor, into the CeLC reversed the behavioral hypersensitivity in a dose-dependent manner. In vitro whole-cell recordings evaluating the change in synaptic transmission found that the frequency as well as amplitude of miniature excitatory postsynaptic currents recorded on CeLC neurons from OIH rats were fundamentally increased and were completely reversed by acutely applied U0126 (10 μM in the recording well). In vivo microinjection of U0126 into the CeLC reversed the spinal long-term potentiation in OIH rats. These results showed that fentanyl-induced hypersensitivity may occur partly through the mechanism of ERK activation and followed by the strengthening of synaptic transmission in CeLC neurons. PERSPECTIVE This study provides evidence that ERK in the laterocapsular division of the CeLC is a key contributor to the development of fentanyl-induced hypersensitivity. Targeting the superspinal central CeLC can inhibit spinal long-term potentiation and alleviate behavioral hyperreflexia induced by fentanyl.
Collapse
|
17
|
de Moura AC, da Silva IRV, Reinaldo G, Dani C, Elsner VR, Giovenardi M. Global Histone H4 Acetylation in the Olfactory Bulb of Lactating Rats with Different Patterns of Maternal Behavior. Cell Mol Neurobiol 2016; 36:1209-13. [PMID: 26620050 PMCID: PMC11482361 DOI: 10.1007/s10571-015-0306-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/17/2015] [Indexed: 12/26/2022]
Abstract
In rats, variations in the levels of neuromodulatory molecules and in the expression of their receptors are observed during pregnancy and postpartum. These changes may contribute to the development and management of maternal behavior. The frequency of licking the pups is used to evaluate maternal care, having mothers with low licking (LL) and high licking (HL) frequencies. Previously, we found that HL had increased levels of transcriptional expression of the receptors for serotonin (HTR1a, HTR1b), estrogen (Erα), dopamine (D1a), and prolactin (Prlr) than LL in the olfactory bulb (OB); however, the molecular mechanisms behind this phenomenon are unknown. Since evidences pointed out that epigenetic marks, which may alter gene expression, are modulated by environmental factors such as exercise, diet, maternal care, and xenobiotic exposure, our objective was to verify the acetylation levels of histone-H4 in the OB of LL and HL rats. Maternal behavior was studied for the first 7 postpartum days. LL (n = 4) and HL (n = 5) mothers were selected according to the behavior of licking their pups. Acetylation levels of histone-H4 were determined using the Global Histone-H4 Acetylation Assay Kit and expressed as ng/mg protein (mean ± SD). Analysis revealed that HL (278.36 ± 68.95) had increased H4 acetylation levels than LL (183.24 ± 73.05; p = 0.045). The enhanced expression of the previously studied receptors in the OB could be related, at least in part, to the hyperacetylation status of histone-H4 here observed. Afterward, the modulation of histone acetylation levels could exert a pivotal role through molecular mechanisms involved in the different patterns of maternal behavior.
Collapse
Affiliation(s)
- Ana Carolina de Moura
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Sarmento Leite 245, Sala 308, Porto Alegre, RS, 90050-170, Brazil.
| | - Ivy Reichert Vital da Silva
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista do IPA, Porto Alegre, RS, Brazil
| | - Gustavo Reinaldo
- Curso de Fisioterapia do Centro Universitário Metodista do IPA, Porto Alegre, RS, Brazil
| | - Caroline Dani
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista do IPA, Porto Alegre, RS, Brazil
| | - Viviane Rostirola Elsner
- Programa de Pós Graduação em Biociências e Reabilitação do Centro Universitário Metodista do IPA, Porto Alegre, RS, Brazil
| | - Márcia Giovenardi
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Sarmento Leite 245, Sala 308, Porto Alegre, RS, 90050-170, Brazil
| |
Collapse
|
18
|
Tao W, Zhou W, Wang Y, Sun T, Wang H, Zhang Z, Jin Y. Histone deacetylase inhibitor-induced emergence of synaptic δ-opioid receptors and behavioral antinociception in persistent neuropathic pain. Neuroscience 2016; 339:54-63. [PMID: 27646288 DOI: 10.1016/j.neuroscience.2016.09.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/31/2016] [Accepted: 09/09/2016] [Indexed: 11/16/2022]
Abstract
The efficacy of opioids in patients with chronic neuropathic pain remains controversial. Although activation of δ-opioid receptors (DORs) in the brainstem reduces inflammation-induced persistent hyperalgesia, it is not effective under persistent neuropathic pain conditions and these clinical problems remain largely unknown. In this study, by using a chronic constriction injury (CCI) of the sciatic nerve in rats, we found that in the brainstem nucleus raphe magnus (NRM), DORs emerged on the surface membrane of central synaptic terminals on day 3 after CCI surgery and disappeared on day 14. Histone deacetylase (HDAC) inhibitors microinjected into the NRM in vivo increased the level of synaptosomal DOR protein and NRM infusion of DOR agonists producing an antinociceptive effect in a nerve growth factor (NGF) signaling-dependent manner. In vitro, in CCI rat slices incubated with HDAC inhibitors, DOR agonists significantly inhibited EPSCs. This effect was blocked by tyrosine receptor kinase A antagonists. Chromatin immunoprecipitation analysis revealed that NRM infusion of HDAC inhibitors in CCI rats increased the level of histone H4 acetylation at Ngf gene promoter regions. NGF was infused into the NRM or incubated CCI rat slices drove DORs to the surface membrane of synaptic terminals. Taken together, epigenetic upregulation of NGF activity by HDAC inhibitors in the NRM promotes the trafficking of DORs to pain-modulating neuronal synapses under neuropathic pain conditions, leading to δ-opioid analgesia. These findings indicate that therapeutic use of DOR agonists combined with HDAC inhibitors might be effective in chronic neuropathic pain managements.
Collapse
Affiliation(s)
- Wenjuan Tao
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Wenjie Zhou
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yuping Wang
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Tingting Sun
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haitao Wang
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhi Zhang
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Yan Jin
- Key Laboratory of Brain Function and Disease of Chinese Academy of Science and Collaborative Innovation Center of Chemistry for Life Sciences, Department of Biophysics and Neurobiology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
19
|
Peregud DI, Yakovlev AA, Stepanichev MY, Onufriev MV, Panchenko LF, Gulyaeva NV. Expression of BDNF and TrkB Phosphorylation in the Rat Frontal Cortex During Morphine Withdrawal are NO Dependent. Cell Mol Neurobiol 2016; 36:839-849. [PMID: 26346883 PMCID: PMC11482428 DOI: 10.1007/s10571-015-0267-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/29/2015] [Indexed: 12/31/2022]
Abstract
Nitric oxide (NO) mediates pharmacological effects of opiates including dependence and abstinence. Modulation of NO synthesis during the induction phase of morphine dependence affects manifestations of morphine withdrawal syndrome, though little is known about mechanisms underlying this phenomenon. Neurotrophic and growth factors are involved in neuronal adaptation during opiate dependence. NO-dependent modulation of morphine dependence may be mediated by changes in expression and activity of neurotrophic and/or growth factors in the brain. Here, we studied the effects of NO synthesis inhibition during the induction phase of morphine dependence on the expression of brain-derived neurotrophic factor (BDNF), glial-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and insulin-like growth factor 1 (IGF1) as well as their receptors in rat brain regions after spontaneous morphine withdrawal in dependent animals. Morphine dependence in rats was induced within 6 days by 12 injections of morphine in increasing doses (10-100 mg/kg), and NO synthase inhibitor L-N(G)-nitroarginine methyl ester (L-NAME) (10 mg/kg) was given 1 h before each morphine injection. The expression of the BDNF, GDNF, NGF, IGF1, and their receptors in the frontal cortex, striatum, hippocampus, and midbrain was assessed 40 h after morphine withdrawal. L-NAME treatment during morphine intoxication resulted in an aggravation of the spontaneous morphine withdrawal severity. Morphine withdrawal was accompanied by upregulation of BDNF, IGF1, and their receptors TrkB and IGF1R, respectively, on the mRNA level in the frontal cortex, and only BDNF in hippocampus and midbrain. L-NAME administration during morphine intoxication decreased abstinence-induced upregulation of these mRNAs in the frontal cortex, hippocampus and midbrain. L-NAME prevented from abstinence-induced elevation of mature but not pro-form of BDNF polypeptide in the frontal cortex. While morphine abstinence did not affect TrkB protein levels as well as its phosphorylation status, inhibition of NO synthesis decreased levels of phosphorylated TrkB after withdrawal. Thus, NO signaling during induction of dependence may be involved in the mechanisms of BDNF expression and processing at abstinence, thereby affecting signaling through TrkB in the frontal cortex.
Collapse
Affiliation(s)
- Danil I Peregud
- Federal State Budgetary Institution "V. Serbsky Federal Medical Research Centre for Psychiatry and Drug Addiction", of the Ministry of Health of the Russian Federation, Moscow, Russia, 119002
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerov Str., Moscow, Russia, 117485
| | - Alexander A Yakovlev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerov Str., Moscow, Russia, 117485
| | - Mikhail Yu Stepanichev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerov Str., Moscow, Russia, 117485
| | - Mikhail V Onufriev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerov Str., Moscow, Russia, 117485
| | - Leonid F Panchenko
- Federal State Budgetary Institution "V. Serbsky Federal Medical Research Centre for Psychiatry and Drug Addiction", of the Ministry of Health of the Russian Federation, Moscow, Russia, 119002
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia, 125315
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5a Butlerov Str., Moscow, Russia, 117485.
| |
Collapse
|
20
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2016; 68:631-700. [PMID: 27343248 PMCID: PMC4931872 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.
Collapse
Affiliation(s)
- Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Catherine M Cahill
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Mark von Zastrow
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Peter W Schiller
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Graciela Pineyro
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| |
Collapse
|
21
|
Epigenetic Manipulation of Brain-derived Neurotrophic Factor Improves Memory Deficiency Induced by Neonatal Anesthesia in Rats. Anesthesiology 2016; 124:624-40. [PMID: 26649423 DOI: 10.1097/aln.0000000000000981] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Although neonatal exposure to anesthetic drugs is associated with memory deficiency in rodent models and possibly in pediatric patients, the underlying mechanisms remain elusive. The authors tested their hypothesis that exposure of the developing brain to anesthesia triggers epigenetic modification, involving the enhanced interaction among transcription factors (histone deacetylase 2, methyl-cytosine-phosphate-guanine-binding protein 2, and DNA methyltransferase 1) in Bdnf promoter region(s) that inhibit brain-derived neurotrophic factor (BDNF) expression, resulting in insufficient drive for local translation of synaptic mRNAs. The authors further hypothesized that noninvasive environmental enrichment (EE) will attenuate anesthesia-induced epigenetic inhibition of BDNF signaling and memory loss in rodent models. METHODS Seven days after birth (P7), neonatal rats were randomly assigned to receive either isoflurane anesthesia for 6 h or sham anesthesia. On P21, pups were weaned, and animals were randomly assigned to EE or a standard cage environment (no EE). Behavioral, molecular, and electrophysiological studies were performed on rats on P65. RESULTS The authors found a substantial reduction of hippocampal BDNF (n = 6 to 7) resulting from the transcriptional factors-mediated epigenetic modification in the promoter region of Bdnf exon IV in rats exposed postnatally to anesthetic drugs. This BDNF reduction led to the insufficient drive for the synthesis of synaptic proteins (n = 6 to 8), thus contributing to the hippocampal synaptic (n = 8 to 11) and cognitive dysfunction (n = 10) induced by neonatal anesthesia. These effects were mitigated by the exposure to an enriched environment. CONCLUSIONS The findings of this study elucidated the epigenetic mechanism underlying memory deficiency induced by neonatal anesthesia and propose EE as a potential therapeutic approach.
Collapse
|
22
|
Dogra S, Sona C, Kumar A, Yadav PN. Epigenetic regulation of G protein coupled receptor signaling and its implications in psychiatric disorders. Int J Biochem Cell Biol 2016; 77:226-39. [PMID: 27046448 DOI: 10.1016/j.biocel.2016.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
G protein-coupled receptors (GPCRs) act as a relay center through which extracellular signals, in the form of neurotransmitters or therapeutics, are converted into an intracellular response, which ultimately shapes the overall response at the tissue and behavioral level. Remarkably in similar ways, epigenetic mechanisms also modulate the expression pattern of a large number of genes in response to the dynamic environment inside and outside of the body, and consequently overall response. Emerging evidences from the pharmacogenomics and preclinical studies clearly suggest that these two distinct mechanisms criss-cross each other in several neurological disorders. At one hand such cross-talks between two distinct mechanisms make disease etiology more challenging to understand, while on the other hand if dealt appropriately, such situations might provide an opportunity to find novel druggable target and strategy for the treatment of complex diseases. In this review article, we have summarized and highlighted the main findings that tie epigenetic mechanisms to GPCR mediated signaling in the pathophysiology of central nervous system (CNS) disorders, including depression, addiction and pain.
Collapse
Affiliation(s)
- Shalini Dogra
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Chandan Sona
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Ajeet Kumar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Prem N Yadav
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India.
| |
Collapse
|
23
|
Persistent pain maintains morphine-seeking behavior after morphine withdrawal through reduced MeCP2 repression of GluA1 in rat central amygdala. J Neurosci 2015; 35:3689-700. [PMID: 25716866 DOI: 10.1523/jneurosci.3453-14.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As long-term opioids are increasingly used for control of chronic pain, how pain affects the rewarding effect of opioids and hence risk of prescription opioid misuse and abuse remains a healthcare concern and a challenging issue in current pain management. In this study, using a rat model of morphine self-administration, we investigated the molecular mechanisms underlying the impact of pain on operant behavior of morphine intake and morphine seeking before and after morphine withdrawal. We found that rats with persistent pain consumed a similar amount of daily morphine to that in control rats without pain, but maintained their level-pressing behavior of morphine seeking after abstinence of morphine at 0.2 mg/kg, whereas this behavior was gradually diminished in control rats. In the central nucleus of amygdala (CeA), a limbic structure critically involved in the affective dimension of pain, proteins of GluA1 subunits of glutamate AMPA receptors were upregulated during morphine withdrawal, and viral knockdown of CeA GluA1 eliminated the morphine-seeking behavior in withdrawn rats of the pain group. Chromatin immunoprecipitation analysis revealed that the methyl CpG-binding protein 2 (MeCP2) was enriched in the promoter region of Gria1 encoding GluA1 and this enrichment was significantly attenuated in withdrawn rats of the pain group. Furthermore, viral overexpression of CeA MeCP2 repressed the GluA1 level and eliminated the maintenance of morphine-seeking behavior after morphine withdrawal. These results suggest direct MeCp2 repression of GluA1 function as a likely mechanism for morphine-seeking behavior maintained by long-lasting affective pain after morphine withdrawal.
Collapse
|
24
|
Bhalla S, Pais G, Tapia M, Gulati A. Endothelin ETA receptor antagonist reverses naloxone-precipitated opioid withdrawal in mice. Can J Physiol Pharmacol 2015; 93:935-44. [PMID: 26440527 DOI: 10.1139/cjpp-2015-0022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Long-term use of opioids for pain management results in rapid development of tolerance and dependence leading to severe withdrawal symptoms. We have previously demonstrated that endothelin-A (ETA) receptor antagonists potentiate opioid analgesia and eliminate analgesic tolerance. This study was designed to investigate the involvement of central ET mechanisms in opioid withdrawal. The effect of intracerebroventricular administration of ETA receptor antagonist BQ123 on morphine and oxycodone withdrawal was determined in male Swiss Webster mice. Opioid tolerance was induced and withdrawal was precipitated by the opioid antagonist naloxone. Expression of ETA and ETB receptors, nerve growth factor (NGF), and vascular endothelial growth factor was determined in the brain using Western blotting. BQ123 pretreatment reversed hypothermia and weight loss during withdrawal. BQ123 also reduced wet shakes, rearing behavior, and jumping behavior. No changes in expression of vascular endothelial growth factor, ETA receptors, and ETB receptors were observed during withdrawal. NGF expression was unaffected in morphine withdrawal but significantly decreased during oxycodone withdrawal. A decrease in NGF expression in oxycodone- but not in morphine-treated mice could be due to mechanistic differences in oxycodone and morphine. It is concluded that ETA receptor antagonists attenuate opioid-induced withdrawal symptoms.
Collapse
Affiliation(s)
- Shaifali Bhalla
- a Department of Pharmaceutical Sciences, Chicago College of Pharmacy, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA
| | - Gwendolyn Pais
- a Department of Pharmaceutical Sciences, Chicago College of Pharmacy, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA
| | - Melissa Tapia
- b Department of Biomedical Sciences, College of Health Sciences, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA
| | - Anil Gulati
- a Department of Pharmaceutical Sciences, Chicago College of Pharmacy, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA
| |
Collapse
|
25
|
Cao S, Qin Y, Chen J, Shen S. Effects of pinacidil on changes to the microenvironment around the incision site, of a skin/muscle incision and retraction, in a rat model of postoperative pain. Mol Med Rep 2015; 12:829-36. [PMID: 25760986 PMCID: PMC4438946 DOI: 10.3892/mmr.2015.3465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 07/23/2014] [Indexed: 12/18/2022] Open
Abstract
The aim of the present study was to evaluate the influence of the microenvironment around an incision site, on peripheral and central sensitization. The effects of pinacidil activation of ATP-sensitive potassium (KATP) channels prior to skin/muscle incision and retraction (SMIR) surgery were assessed. A total of 24 male Sprague Dawley rats were randomly assigned to four groups: Control, sham (incision operation), SMIR (incision plus retraction 1 h after the skin/muscle incision) and pinacidil (SMIR plus pinacidil). The rats in the pinacidil group were intraperitoneally injected with pinacidil prior to the SMIR procedure. The mechanical withdrawal threshold (MWT) was determined at each time point. The microvessel density (MVD) value was determined by immunohistochemistry, and western blotting was performed to analyze the relative protein expression levels of nerve growth factor (NGF), glucose transporter protein-1 (GLUT1) and C-jun N-terminal kinases. There was a significant reduction in the levels of MVD, GLUT1 and MWT following SMIR surgery as compared with the incision alone, and a significant increase in the NGF protein expression levels. In the SMIR group, the MVD value was significantly increased seven days after surgery, as compared with three days after surgery. Additionally, intraperitoneal administration of pinacidil prior to the SMIR surgery inhibited the SMIR-induced reduction in MWT and MVD and attenuated the SMIR-induced GLUT1 reduction. The results of the present study suggest that the microenvironment around an incision site may affect the development of peripheral and central sensitization. In addition, pinacidil had an inhibitory effect on the formation of the inflammatory microenvironment around the incision site through activation of KATP channels, thereby inhibiting peripheral and central sensitization.
Collapse
Affiliation(s)
- Su Cao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yinbin Qin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Junjie Chen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Shiren Shen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
26
|
Abstract
A limbic brain area, the amygdala plays a key role in emotional responses and affective states and disorders such as learned fear, anxiety, and depression. The amygdala has also emerged as an important brain center for the emotional-affective dimension of pain and for pain modulation. Hyperactivity in the laterocapsular division of the central nucleus of the amygdala (CeLC, also termed the "nociceptive amygdala") accounts for pain-related emotional responses and anxiety-like behavior. Abnormally enhanced output from the CeLC is the consequence of an imbalance between excitatory and inhibitory mechanisms. Impaired inhibitory control mediated by a cluster of GABAergic interneurons in the intercalated cell masses (ITC) allows the development of glutamate- and neuropeptide-driven synaptic plasticity of excitatory inputs from the brainstem (parabrachial area) and from the lateral-basolateral amygdala network (LA-BLA, site of integration of polymodal sensory information). BLA hyperactivity also generates abnormally enhanced feedforward inhibition of principal cells in the medial prefrontal cortex (mPFC), a limbic cortical area that is strongly interconnected with the amygdala. Pain-related mPFC deactivation results in cognitive deficits and failure to engage cortically driven ITC-mediated inhibitory control of amygdala processing. Impaired cortical control allows the uncontrolled persistence of amygdala pain mechanisms.
Collapse
Affiliation(s)
- Volker Neugebauer
- Department of Pharmacology and Neuroscience, Center for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430-6592, USA,
| |
Collapse
|
27
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
28
|
Nerve growth factor sensitizes adult sympathetic neurons to the proinflammatory peptide bradykinin. J Neurosci 2014; 34:11959-71. [PMID: 25186743 DOI: 10.1523/jneurosci.1536-14.2014] [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] [Indexed: 12/12/2022] Open
Abstract
Levels of nerve growth factor (NGF) are elevated in inflamed tissues. In sensory neurons, increases in NGF augment neuronal sensitivity (sensitization) to noxious stimuli. Here, we hypothesized that NGF also sensitizes sympathetic neurons to proinflammatory stimuli. We cultured superior cervical ganglion (SCG) neurons from adult male Sprague Dawley rats with or without added NGF and compared their responsiveness to bradykinin, a proinflammatory peptide. The NGF-cultured neurons exhibited significant depolarization, bursts of action potentials, and Ca(2+) elevations after bradykinin application, whereas neurons cultured without NGF showed only slight changes in membrane potential and cytoplasmic Ca(2+) levels. The NGF effect, which requires trkA receptors, takes hours to develop and days to reverse. We addressed the ionic mechanisms underlying this sensitization. NGF did not alter bradykinin-induced M-current inhibition or phosphatidylinositol 4,5-bisphosphate hydrolysis. Maxi-K channel-mediated current evoked by depolarizations was reduced by 50% by culturing neurons in NGF. Application of iberiotoxin or paxilline, blockers of Maxi-K channels, mimicked NGF treatment and sensitized neurons to bradykinin application. A calcium channel blocker also mimicked NGF treatment. We found that NGF reduces Maxi-K channel opening by decreasing the activity of nifedipine-sensitive calcium channels. In conclusion, culture in NGF reduces the activity of L-type calcium channels, and secondarily, the calcium-sensitive activity of Maxi-K channels, rendering sympathetic neurons electrically hyper-responsive to bradykinin.
Collapse
|
29
|
Tao W, Chen Q, Zhou W, Wang Y, Wang L, Zhang Z. Persistent inflammation-induced up-regulation of brain-derived neurotrophic factor (BDNF) promotes synaptic delivery of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluA1 subunits in descending pain modulatory circuits. J Biol Chem 2014; 289:22196-204. [PMID: 24966334 PMCID: PMC4139232 DOI: 10.1074/jbc.m114.580381] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/16/2014] [Indexed: 11/06/2022] Open
Abstract
The enhanced AMPA receptor phosphorylation at GluA1 serine 831 sites in the central pain-modulating system plays a pivotal role in descending pain facilitation after inflammation, but the underlying mechanisms remain unclear. We show here that, in the rat brain stem, in the nucleus raphe magnus, which is a critical relay in the descending pain-modulating system of the brain, persistent inflammatory pain induced by complete Freund adjuvant (CFA) can enhance AMPA receptor-mediated excitatory postsynaptic currents and the GluA2-lacking AMPA receptor-mediated rectification index. Western blot analysis showed an increase in GluA1 phosphorylation at Ser-831 but not at Ser-845. This was accompanied by an increase in distribution of the synaptic GluA1 subunit. In parallel, the level of histone H3 acetylation at bdnf gene promoter regions was reduced significantly 3 days after CFA injection, as indicated by ChIP assays. This was correlated with an increase in BDNF mRNA levels and BDNF protein levels. Sequestering endogenous extracellular BDNF with TrkB-IgG in the nucleus raphe magnus decreased AMPA receptor-mediated synaptic transmission and GluA1 phosphorylation at Ser-831 3 days after CFA injection. Under the same conditions, blockade of TrkB receptor functions, phospholipase C, or PKC impaired GluA1 phosphorylation at Ser-831 and decreased excitatory postsynaptic currents mediated by GluA2-lacking AMPA receptors. Taken together, these results suggest that epigenetic up-regulation of BDNF by peripheral inflammation induces GluR1 phosphorylation at Ser-831 sites through activation of the phospholipase C-PKC signaling cascade, leading to the trafficking of GluA1 to pain-modulating neuronal synapses.
Collapse
Affiliation(s)
- Wenjuan Tao
- From the Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China and the Department of Pharmacy, Anhui College of Traditional Chinese Medicine, Wuhu, Anhui 241000, China
| | - Quan Chen
- From the Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China and
| | - Wenjie Zhou
- From the Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China and
| | - Yunping Wang
- From the Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China and
| | - Lu Wang
- From the Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China and
| | - Zhi Zhang
- From the Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China and
| |
Collapse
|
30
|
Sun Y, Sahbaie P, Liang D, Li W, Clark JD. Opioids enhance CXCL1 expression and function after incision in mice. THE JOURNAL OF PAIN 2014; 15:856-66. [PMID: 24887006 PMCID: PMC4131856 DOI: 10.1016/j.jpain.2014.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/29/2014] [Accepted: 05/20/2014] [Indexed: 12/20/2022]
Abstract
UNLABELLED Chronic opioid consumption increases postoperative pain. Epigenetic changes related to chronic opioid use and surgical incision may be partially responsible for this enhancement. The CXCL1/CXCR2 signaling pathway, implicated in several pain models, is known to be epigenetically regulated via histone acetylation. The current study was designed to investigate the role of CXCL1/CXCR2 signaling in opioid-enhanced incisional sensitization and to elucidate the possible epigenetic mechanism underlying CXCL1/CXCR2 pathway-mediated regulation of nociceptive sensitization in mice. Chronic morphine treatment generated mechanical and thermal nociceptive sensitization and also significantly exacerbated incision-induced mechanical allodynia. Peripheral but not central messenger RNA levels of CXCL1 and CXCR2 were increased after incision. The source of peripheral CXCL1 appeared to be wound area neutrophils. Histone H3 subunit acetylated at the lysine 9 position (AcH3K9) was increased in infiltrating dermal neutrophils after incision and was further increased in mice with chronic morphine treatment. The association of AcH3K9 with the promoter region of CXCL1 was enhanced in mice after chronic morphine treatment. The increase in CXCL1 near wounds caused by chronic morphine pretreatment was mimicked by pharmacologic inhibition of histone deacetylation. Finally, local injection of CXCL1 induced mechanical sensitivity in naive mice, whereas blocking CXCR2 reversed mechanical hypersensitivity after hind paw incision. PERSPECTIVE Peripheral CXCL1/CXCR2 signaling helps to control nociceptive sensitization after incision, and epigenetic regulation of CXCL1 expression explains in part opioid-enhanced incisional allodynia in mice. These results suggest that targeting CXCL1/CXCR2 signaling may be useful in treating nociceptive sensitization, particularly for postoperative pain in chronic opioid-consuming patients.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/pharmacology
- Animals
- Chemokine CXCL1/genetics
- Chemokine CXCL1/metabolism
- Disease Models, Animal
- Drug Administration Schedule
- Gene Expression Regulation/drug effects
- Hyperalgesia/drug therapy
- Hyperalgesia/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Morphine/administration & dosage
- Neutrophils/drug effects
- Neutrophils/metabolism
- Pain Measurement
- Pain Threshold/drug effects
- Pain, Postoperative/drug therapy
- Pain, Postoperative/metabolism
- Phenylurea Compounds/administration & dosage
- Phenylurea Compounds/pharmacology
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Time Factors
- Wounds, Penetrating/complications
- Wounds, Penetrating/drug therapy
Collapse
Affiliation(s)
- Yuan Sun
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Peyman Sahbaie
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - DeYong Liang
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Wenwu Li
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California
| | - J David Clark
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, California; Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, California.
| |
Collapse
|
31
|
Zhang Z, Tao W, Hou YY, Wang W, Lu YG, Pan ZZ. Persistent pain facilitates response to morphine reward by downregulation of central amygdala GABAergic function. Neuropsychopharmacology 2014; 39:2263-71. [PMID: 24686896 PMCID: PMC4104345 DOI: 10.1038/npp.2014.77] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/21/2014] [Accepted: 03/25/2014] [Indexed: 01/11/2023]
Abstract
Opioid-based analgesics are widely used for treating chronic pain, but opioids are highly addictive when repeatedly used because of their strong rewarding effects. In recent years, abuse of prescription opioids has dramatically increased, including incidences of misuse of opioid drugs prescribed for pain control. Despite this issue in current clinical pain management, it remains unknown how pain influences the abuse liability of prescription opioids. Pain as aversive experience may affect opioid reward of positive emotion through common brain sites involved in emotion processing. In this study, on a rat model of chronic pain, we determined how persistent pain altered behavioral responses to morphine reward measured by the paradigm of unbiased conditioned place preference (CPP), focusing on GABAergic synaptic activity in neurons of the central nucleus of the amygdala (CeA), an important brain region for emotional processing of both pain and reward. We found that pain reduced the minimum number of morphine-conditioning sessions required for inducing CPP behavior. Both pain and morphine conditioning that elicited CPP inhibited GABA synaptic transmission in CeA neurons. Pharmacological activation of CeA GABAA receptors reduced the pain and inhibited CPP induced both by an effective dose of morphine and by a sub-threshold dose of morphine under pain condition. Furthermore, inhibition of CeA GABAA receptors mimicked the pain effect, rendering the sub-threshold dose of morphine effective in CPP induction. These findings suggest that pain facilitates behavioral responses to morphine reward by predisposing the inhibitory GABA function in the CeA circuitry involved in the behavior of opioid reward.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA,Department of Neurobiology and Biophysics, Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, China,Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei 230027, China. E-mail:
| | - Wenjuan Tao
- Department of Neurobiology and Biophysics, Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Yuan-Yuan Hou
- Department of Anesthesiology and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Wang
- Department of Anesthesiology and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yun-Gang Lu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhizhong Z Pan
- Department of Anesthesiology and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA,Department of Anesthesiology and Pain Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 110, Houston, TX 77030, USA, Tel: +713 792 5559, Fax: +713 745 3040, E-mail:
| |
Collapse
|
32
|
Klenowski P, Morgan M, Bartlett SE. The role of δ-opioid receptors in learning and memory underlying the development of addiction. Br J Pharmacol 2014; 172:297-310. [PMID: 24641428 DOI: 10.1111/bph.12618] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/10/2014] [Accepted: 01/19/2014] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED Opioids are important endogenous ligands that exist in both invertebrates and vertebrates and signal by activation of opioid receptors to produce analgesia and reward or pleasure. The μ-opioid receptor is the best known of the opioid receptors and mediates the acute analgesic effects of opiates, while the δ-opioid receptor (DOR) has been less well studied and has been linked to effects that follow from chronic use of opiates such as stress, inflammation and anxiety. Recently, DORs have been shown to play an essential role in emotions and increasing evidence points to a role in learning actions and outcomes. The process of learning and memory in addiction has been proposed to involve strengthening of specific brain circuits when a drug is paired with a context or environment. The DOR is highly expressed in the hippocampus, amygdala, striatum and other basal ganglia structures known to participate in learning and memory. In this review, we will focus on the role of the DOR and its potential role in learning and memory underlying the development of addiction. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- Paul Klenowski
- Translational Research Institute, Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | | | | |
Collapse
|
33
|
Wang Z, Yan P, Hui T, Zhang J. Epigenetic upregulation of PSD-95 contributes to the rewarding behavior by morphine conditioning. Eur J Pharmacol 2014; 732:123-9. [PMID: 24704371 DOI: 10.1016/j.ejphar.2014.03.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 03/04/2014] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
Abstract
Abuse of opiates, including morphine, induced remarkable synaptic adaptation in several brain regions including ventral tegmental area (VTA), which underlay the induction and maintenance of opioid dependence and addiction. Scaffolding protein postsynaptic density protein 95 (PSD-95) is critically involved in the glutamatergic synaptic maturation and plasticity in the central neurons. The present study revealed a significantly increased mRNA and protein expression of PSD-95 in the VTA of the rats conditioned with morphine. The further chromatin immunoprecipitation study found an increased histone H3 acetylation in the promoter region of Dlg4. An upregulation of expression of phosphorylated cAMP response element-binding protein (pCREB) and the occupancy of pCREB in the Dlg4 promoter region were shown in the VTA of the morphine-conditioned rats. Inhibition of pCREB activity significantly decreased the histone H3 acetylation in Dlg4 promoter region, PSD-95 upregulation, enhancement of glutamatergic strength and the preference to morphine-paired chamber in the rats with morphine conditioning. These results suggested that CREB-mediated epigenetic upregulation of PSD-95 critically contributed to the enhanced glutamatergic transmission and rewarding behavior induced by morphine conditioning.
Collapse
Affiliation(s)
- Ziwen Wang
- Department of Preventive Medicine, Third Military Medical University, Chongqing 400038, China; Department of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Pingao Yan
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tianli Hui
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiqiang Zhang
- Department of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
34
|
Bie B, Wu J, Yang H, Xu JJ, Brown DL, Naguib M. Epigenetic suppression of neuroligin 1 underlies amyloid-induced memory deficiency. Nat Neurosci 2014; 17:223-31. [PMID: 24441681 DOI: 10.1038/nn.3618] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/27/2013] [Indexed: 12/19/2022]
Abstract
Amyloid-induced microglial activation and neuroinflammation impair central synapses and memory function, although the mechanism remains unclear. Neuroligin 1 (NLGN1), a postsynaptic protein found in central excitatory synapses, governs excitatory synaptic efficacy and plasticity in the brain. Here we found, in rodents, that amyloid fibril-induced neuroinflammation enhanced the interaction between histone deacetylase 2 and methyl-CpG-binding protein 2, leading to suppressed histone H3 acetylation and enhanced cytosine methylation in the Nlgn1 promoter region and decreased NLGN1 expression, underlying amyloid-induced memory deficiency. Manipulation of microglia-associated neuroinflammation modulated the epigenetic modification of the Nlgn1 promoter, hippocampal glutamatergic transmission and memory function. These findings link neuroinflammation, synaptic efficacy and memory, thus providing insight into the pathogenesis of amyloid-associated diseases.
Collapse
Affiliation(s)
- Bihua Bie
- 1] Department of General Anesthesiology, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio, USA. [2]
| | - Jiang Wu
- 1] Department of General Anesthesiology, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio, USA. [2]
| | - Hui Yang
- Department of General Anesthesiology, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jijun J Xu
- Department of General Anesthesiology, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - David L Brown
- Department of General Anesthesiology, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mohamed Naguib
- Department of General Anesthesiology, Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Zhang Z, Wang X, Wang W, Lu YG, Pan ZZ. Brain-derived neurotrophic factor-mediated downregulation of brainstem K+-Cl- cotransporter and cell-type-specific GABA impairment for activation of descending pain facilitation. Mol Pharmacol 2013; 84:511-20. [PMID: 23847084 PMCID: PMC3781381 DOI: 10.1124/mol.113.086496] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 07/11/2013] [Indexed: 11/22/2022] Open
Abstract
Chronic pain is thought to be partly caused by a loss of GABAergic inhibition and resultant neuronal hyperactivation in the central pain-modulating system, but the underlying mechanisms for pain-modulating neurons in the brain are unclear. In this study, we investigated the cellular mechanisms for activation of brainstem descending pain facilitation in rats under persistent pain conditions. In the nucleus raphe magnus (NRM), a critical relay in the brain's descending pain-modulating system, persistent inflammatory pain induced by complete Freund's adjuvant decreased the protein level of K(+)-Cl(-) cotransporter (KCC2) in both total and synaptosomal preparations. Persistent pain also shifted the equilibrium potential of GABAergic inhibitory postsynaptic current (EIPSC) to a more positive level and increased the firing of evoked action potentials selectively in μ-opioid receptor (MOR)-expressing NRM neurons, but not in MOR-lacking NRM neurons. Microinjection of brain-derived neurotrophic factor (BDNF) into the NRM inhibited the KCC2 protein level in the NRM, and both BDNF administration and KCC2 inhibition by furosemide mimicked the pain-induced effects on EIPSC and excitability in MOR-expressing neurons. Furthermore, inhibiting BDNF signaling by NRM infusion of tyrosine receptor kinase B-IgG or blocking KCC2 with furosemide prevented these pain effects in MOR-expressing neurons. These findings demonstrate a cellular mechanism by which the hyperactivity of NRM MOR-expressing neurons, presumably responsible for descending pain facilitation, contributes to pain sensitization through the signaling cascade of BDNF-KCC2-GABA impairment in the development of chronic pain.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Pain Medicine, University of Texas, MD Anderson Cancer Center, Houston, Texas (Z.Z., W.W., Z.Z.P.); Department of Neurobiology, University of Science and Technology of China, Hefei, People's Republic of China (Z.Z., X.W.); and Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas (Y.-G.L.)
| | | | | | | | | |
Collapse
|
36
|
Zhu F, Yan CX, Wen YC, Wang J, Bi J, Zhao YL, Wei L, Gao CG, Jia W, Li SB. Dopamine D1 receptor gene variation modulates opioid dependence risk by affecting transition to addiction. PLoS One 2013; 8:e70805. [PMID: 23976958 PMCID: PMC3745389 DOI: 10.1371/journal.pone.0070805] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/21/2013] [Indexed: 01/25/2023] Open
Abstract
Dopamine D1 receptor (DRD1) modulates opioid reinforcement, reward, and opioid-induced neuroadaptation. We propose that DRD1 polymorphism affects susceptibility to opioid dependence (OD), the efficiency of transition to OD, and opioid-induced pleasure response. We analyzed potential association between seven DRD1 polymorphisms with the following traits: duration of transition from the first use to dependence (DTFUD), subjective pleasure responses to opioid on first use and post-dependence use, and OD risk in 425 Chinese with OD and 514 healthy controls. DTFUD and level of pleasure responses were examined using a semi-structured interview. The DTFUD of opioid addicts ranged from 5 days to 11 years. Most addicts (64.0%) reported non-comfortable response upon first opioid use, while after dependence, most addicts (53.0%) felt strong opioid-induced pleasure. Survival analysis revealed a correlation of prolonged DTFUD with the minor allele-carrying genotypes of DRD1 rs4532 (hazard ratios (HR) = 0.694; p = 0.001) and rs686 (HR = 0.681, p = 0.0003). Binary logistic regression indicated that rs10063995 GT genotype (vs. GG+TT, OR = 0.261) could predict decreased pleasure response to first-time use and the minor alleles of rs686 (OR = 0.535) and rs4532 (OR = 0.537) could predict decreased post-dependence pleasure. Moreover, rs686 minor allele was associated with a decreased risk for rapid transition from initial use to dependence (DTFUD≤30 days; OR = 0.603) or post-dependence euphoria (OR = 0.603) relative to major allele. In conclusion, DRD1 rs686 minor allele decreases the OD risk by prolonging the transition to dependence and attenuating opioid-induced pleasure in Chinese.
Collapse
Affiliation(s)
- Feng Zhu
- College of Medicine and Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Health Ministry for Forensic Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Education Ministry for Environment and Genes Related to Diseases, Xi'an, Shaanxi, People's Republic of China
| | - Chun-xia Yan
- College of Medicine and Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Health Ministry for Forensic Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Education Ministry for Environment and Genes Related to Diseases, Xi'an, Shaanxi, People's Republic of China
| | - Yi-chong Wen
- College of Medicine and Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Health Ministry for Forensic Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Education Ministry for Environment and Genes Related to Diseases, Xi'an, Shaanxi, People's Republic of China
| | - Jiayin Wang
- The Genome Institute, Washington University, Saint Louis, Missouri, United States of America
| | - Jinbo Bi
- The Genome Institute, Washington University, Saint Louis, Missouri, United States of America
- Department of Community Medicine and Health Care, School of Medicine University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Ya-ling Zhao
- Key Laboratory of the Education Ministry for Environment and Genes Related to Diseases, Xi'an, Shaanxi, People's Republic of China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Lai Wei
- College of Medicine and Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Health Ministry for Forensic Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Education Ministry for Environment and Genes Related to Diseases, Xi'an, Shaanxi, People's Republic of China
| | - Cheng-ge Gao
- Department of Psychiatry, First Affiliated Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Wei Jia
- Methadone Maintenance Therapy Clinic, Xi'an Mental Health Center, Xi'an, Shaanxi, People's Republic of China
| | - Sheng-bin Li
- College of Medicine and Forensics, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Health Ministry for Forensic Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of the Education Ministry for Environment and Genes Related to Diseases, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
37
|
Wu J, Bie B, Yang H, Xu JJ, Brown DL, Naguib M. Suppression of central chemokine fractalkine receptor signaling alleviates amyloid-induced memory deficiency. Neurobiol Aging 2013; 34:2843-52. [PMID: 23855980 DOI: 10.1016/j.neurobiolaging.2013.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 05/18/2013] [Accepted: 06/08/2013] [Indexed: 10/26/2022]
Abstract
The abnormal accumulation of amyloid fibrils in the brain is pathognomonic of Alzheimer's disease. Amyloid fibrils induce significant neuroinflammation characterized by the activation of microglia and impairment of synaptic plasticity in the brain that eventually leads to cognitive decline. Chemokine fractalkine receptor (CX3CR1) is primarily located in the microglia in the brain and its role in the amyloid fibril-induced neuroinflammation and memory deficiency remains debated. We found that bilateral microinjection of amyloid beta (Aβ)1-40 fibrils into the hippocampal CA1 area of rats resulted in significant upregulation of CX3CR1 messenger RNA (mRNA) and protein expression (via increasing histone H3 acetylation in the Cx3cr1 promoter region), synaptic dysfunction, and cognitive impairment, compared with the control group. Suppressing CX3CR1 signaling with CX3CR1 small interfering RNA in rats injected with Aβ1-40 fibrils blunted Aβ1-40-induced CX3CR1 upregulation, microglial activation, interleukin-1β expression, restored basal glutamatergic strength and electric stimuli-induced long-term potentiation, and cognitive capacities. These findings suggest that activation of CX3CR1 plays an important role in the neuroinflammatory response and Aβ-induced neuroinflammation and neurotoxicity.
Collapse
Affiliation(s)
- Jiang Wu
- Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, USA; College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | | | | | | | | | | |
Collapse
|
38
|
HDAC inhibitors attenuate the development of hypersensitivity in models of neuropathic pain. Pain 2013; 154:1668-1679. [PMID: 23693161 PMCID: PMC3763368 DOI: 10.1016/j.pain.2013.05.021] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/23/2013] [Accepted: 05/03/2013] [Indexed: 12/20/2022]
Abstract
Histone deacetylase inhibitors (HDACIs) interfere with the epigenetic process of histone acetylation and are known to have analgesic properties in models of chronic inflammatory pain. The aim of this study was to determine whether these compounds could also affect neuropathic pain. Different class I HDACIs were delivered intrathecally into rat spinal cord in models of traumatic nerve injury and antiretroviral drug–induced peripheral neuropathy (stavudine, d4T). Mechanical and thermal hypersensitivity was attenuated by 40% to 50% as a result of HDACI treatment, but only if started before any insult. The drugs globally increased histone acetylation in the spinal cord, but appeared to have no measurable effects in relevant dorsal root ganglia in this treatment paradigm, suggesting that any potential mechanism should be sought in the central nervous system. Microarray analysis of dorsal cord RNA revealed the signature of the specific compound used (MS-275) and suggested that its main effect was mediated through HDAC1. Taken together, these data support a role for histone acetylation in the emergence of neuropathic pain.
Collapse
|
39
|
Abstract
Harmful excessive use of alcohol has a severe impact on society and it remains one of the major causes of morbidity and mortality in the population. However, mechanisms that underlie excessive alcohol consumption are still poorly understood, and thus available medications for alcohol use disorders are limited. Here, we report that changing the level of chromatin condensation by affecting DNA methylation or histone acetylation limits excessive alcohol drinking and seeking behaviors in rodents. Specifically, we show that decreasing DNA methylation by inhibiting the activity of DNA methyltransferase (DNMT) with systemic administration of the FDA-approved drug, 5-azacitidine (5-AzaC) prevents excessive alcohol use in mice. Similarly, we find that increasing histone acetylation via systemic treatment with several histone deacetylase (HDAC) inhibitors reduces mice binge-like alcohol drinking. We further report that systemic administration of the FDA-approved HDAC inhibitor, SAHA, inhibits the motivation of rats to seek alcohol. Importantly, the actions of both DNMT and HDAC inhibitors are specific for alcohol, as no changes in saccharin or sucrose intake were observed. In line with these behavioral findings, we demonstrate that excessive alcohol drinking increases DNMT1 levels and reduces histone H4 acetylation in the nucleus accumbens (NAc) of rodents. Together, our findings illustrate that DNA methylation and histone acetylation control the level of excessive alcohol drinking and seeking behaviors in preclinical rodent models. Our study therefore highlights the possibility that DNMT and HDAC inhibitors can be used to treat harmful alcohol abuse.
Collapse
|