1
|
Zhao X, Hu A, Wang Y, Zhao T, Xiang X. Paraventricular thalamus to nucleus accumbens circuit activation decreases long-term relapse of alcohol-seeking behaviour in male mice. Pharmacol Biochem Behav 2024; 237:173726. [PMID: 38360104 DOI: 10.1016/j.pbb.2024.173726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/23/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Some studies have highlighted the crucial role of aversion in addiction treatment. The pathway from the anterior paraventricular thalamus (PVT) to the shell of the nucleus accumbens (NAc) has been reported as an essential regulatory pathway for processing aversion and is also closely associated with substance addiction. However, its impact on alcohol addiction has been relatively underexplored. Therefore, this study focused on the role of the PVT-NAc pathway in the formation and relapse of alcohol addiction-like behaviour, offering a new perspective on the mechanisms of alcohol addiction. RESULTS The chemogenetic inhibition of the PVT-NAc pathway in male mice resulted in a notable decrease in the establishment of ethanol-induced conditioned place aversion (CPA), and NAc-projecting PVT neurons were recruited due to aversive effects. Conversely, activation of the PVT-NAc pathway considerably impeded the formation of ethanol-induced conditioned place preference (CPP). Furthermore, during the memory reconsolidation phase, activation of this pathway effectively disrupted the animals' preference for alcohol-associated contexts. Whether it was administered urgently 24 h later or after a long-term withdrawal of 10 days, a low dose of alcohol could still not induce the reinstatement of ethanol-induced CPP. CONCLUSIONS Our results demonstrated PVT-NAc circuit processing aversion, which may be one of the neurobiological mechanisms underlying aversive counterconditioning, and highlighted potential targets for inhibiting the development of alcohol addiction-like behaviour and relapse after long-term withdrawal.
Collapse
Affiliation(s)
- Xiaoxi Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Aqian Hu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yanyan Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Tianshu Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xiaojun Xiang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
2
|
Fu Y, Li W, Mai Y, Guan J, Ding R, Hou J, Chen B, Cao G, Sun S, Tang Y, Fu R. Association between RMTg Neuropeptide Genes and Negative Effect during Alcohol Withdrawal in Mice. Int J Mol Sci 2024; 25:2933. [PMID: 38474180 DOI: 10.3390/ijms25052933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Alcohol use disorders (AUDs) frequently co-occur with negative mood disorders, such as anxiety and depression, exacerbating relapse through dopaminergic dysfunction. Stress-related neuropeptides play a crucial role in AUD pathophysiology by modulating dopamine (DA) function. The rostromedial tegmental nucleus (RMTg), which inhibits midbrain dopamine neurons and signals aversion, has been shown to increase ethanol consumption and negative emotional states during abstinence. Despite some stress-related neuropeptides acting through the RMTg to affect addiction behaviors, their specific roles in alcohol-induced contexts remain underexplored. This study utilized an intermittent voluntary drinking model in mice to induce negative effect behavior 24 h into ethanol (EtOH) abstinence (post-EtOH). It examined changes in pro-stress (Pnoc, Oxt, Npy) and anti-stress (Crf, Pomc, Avp, Orx, Pdyn) neuropeptide-coding genes and analyzed their correlations with aversive behaviors. We observed that adult male C57BL/6J mice displayed evident anxiety, anhedonia, and depression-like symptoms at 24 h post-EtOH. The laser-capture microdissection technique, coupled with or without retrograde tracing, was used to harvest total ventral tegmental area (VTA)-projecting neurons or the intact RMTg area. The findings revealed that post-EtOH consistently reduced Pnoc and Orx levels while elevating Crf levels in these neuronal populations. Notably, RMTg Pnoc and Npy levels counteracted ethanol consumption and depression severity, while Crf levels were indicative of the mice's anxiety levels. Together, these results underscore the potential role of stress-related neuropeptides in the RMTg in regulating the negative emotions related to AUDs, offering novel insights for future research.
Collapse
Affiliation(s)
- Yixin Fu
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Yunlin Mai
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Junhao Guan
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Ruxuan Ding
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Jiawei Hou
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Bingqing Chen
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Guoxin Cao
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Shizhu Sun
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Ying Tang
- Clinical Skills Training Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Sun Yat-Sen University, Shenzhen 518106, China
| |
Collapse
|
3
|
Becker HC, Lopez MF, King CE, Griffin WC. Oxytocin Reduces Sensitized Stress-Induced Alcohol Relapse in a Model of Posttraumatic Stress Disorder and Alcohol Use Disorder Comorbidity. Biol Psychiatry 2023; 94:215-225. [PMID: 36822933 PMCID: PMC10247903 DOI: 10.1016/j.biopsych.2022.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND There is high comorbidity of posttraumatic stress disorder (PTSD) and alcohol use disorder with few effective treatment options. Animal models of PTSD have shown increases in alcohol drinking, but effects of stress history on subsequent vulnerability to alcohol relapse have not been examined. Here we present a mouse model of PTSD involving chronic multimodal stress exposure that resulted in long-lasting sensitization to stress-induced alcohol relapse, and this sensitized stress response was blocked by oxytocin (OT) administration. METHODS Male and female mice trained to self-administer alcohol were exposed to predator odor (TMT) + yohimbine over 5 consecutive days or left undisturbed. After reestablishing stable alcohol responding/intake, mice were tested under extinction conditions, and then all mice were exposed to TMT or context cues previously associated with TMT before a reinstatement test session. Separate studies examined messenger RNA expression of Oxt and Oxtr in hypothalamus following chronic stress exposure. A final study examined the effects of systemic administration of OT on stress-induced alcohol relapse in mice with and without a history of chronic stress experience. RESULTS Chronic stress exposure produced long-lasting sensitization to subsequent stress-induced alcohol relapse that also generalized to stress-related context cues and transcriptional changes in hypothalamic OT system. OT injected before the reinstatement test session completely blocked the sensitized stress-induced alcohol relapse effect. CONCLUSIONS Collectively, these results provide support for the therapeutic potential of OT, along with highlighting the value of utilizing this model in evaluating other pharmacological interventions for treatment of PTSD/alcohol use disorder comorbidity.
Collapse
Affiliation(s)
- Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina; Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, South Carolina.
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Courtney E King
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
4
|
Decker Ramirez EB, Arnold ME, McConnell KT, Solomon MG, Amico KN, Schank JR. The effects of lipopolysaccharide exposure on social interaction, cytokine expression, and alcohol consumption in male and female mice. Physiol Behav 2023; 265:114159. [PMID: 36931488 PMCID: PMC10121933 DOI: 10.1016/j.physbeh.2023.114159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/10/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Much recent research has demonstrated a role of inflammatory pathways in depressive-like behavior and excess alcohol consumption. Lipopolysaccharide (LPS) is a cell wall component of gram-negative bacteria that can be used to trigger a strong inflammatory response in rodents in a preclinical research setting to study the mechanisms behind this relationship. In our study, we exposed male and female mice to LPS and assessed depressive-like behavior using the social interaction (SI) test, alcohol consumption in the two-bottle choice procedure, and expression of inflammatory mediators using quantitative PCR. We found that LPS administration decreased SI in female mice but had no significant impact on male mice when assessed 24 h after injection. LPS resulted in increased proinflammatory cytokine expression in both male and female mice; however, some aspects of the cytokine upregulation observed was greater in female mice as compared to males. A separate cohort of male and female mice underwent drinking for 12 days before receiving a saline or LPS injection, which we found to increase alcohol intake in both males and females. We have previously observed a role of the neurokinin-1 receptor (NK1R) in escalated alcohol intake, and in the inflammatory and behavioral response to LPS. The NK1R is the endogenous target of the neuropeptide SP, and this system has wide ranging roles in depression, anxiety, drug/alcohol seeking, pain, and inflammation. Thus, we administered a NK1R antagonist prior to alcohol access. This treatment reduced escalated alcohol consumption in female mice treated with LPS but did not affect drinking in males. Taken together, these results indicate that females are more sensitive to some physiological and behavioral effects of LPS administration, but that LPS escalates alcohol consumption in both sexes. Furthermore, NK1R antagonism can reduce alcohol consumption that is escalated by LPS treatment, in line with our previous findings.
Collapse
Affiliation(s)
- E B Decker Ramirez
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - M E Arnold
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - K T McConnell
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - M G Solomon
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - K N Amico
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - J R Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA.
| |
Collapse
|
5
|
Li W, Ren Z, Tang Y, Fu Y, Sun S, Ding R, Hou J, Mai Y, Zhan B, Zhu Y, Zuo W, Ye JH, Fu R. Rostromedial tegmental nucleus nociceptin/orphanin FQ (N/OFQ) signaling regulates anxiety- and depression-like behaviors in alcohol withdrawn rats. Neuropsychopharmacology 2023; 48:908-919. [PMID: 36329156 PMCID: PMC10156713 DOI: 10.1038/s41386-022-01482-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/24/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Recent studies indicate that stimulation of the rostromedial tegmental nucleus (RMTg) can drive a negative affective state and that nociceptin/orphanin FQ (N/OFQ) may play a role in affective disorders and drug addiction. The N/OFQ precursor prepronociceptin encoding genes Pnoc are situated in RMTg neurons. To determine whether N/OFQ signaling contributes to the changes in both behavior phenotypes and RMTg activity of alcohol withdrawn (Post-EtOH) rats, we trained adult male Long-Evans rats, randomly assigned into the ethanol and Naïve groups to consume either 20% ethanol or water-only under an intermittent-access procedure. Using the fluorescence in situ hybridization technique combined with retrograde tracing, we show that the ventral tegmental area projecting RMTg neurons express Pnoc and nociceptin opioid peptide (NOP) receptors encoding gene Oprl1. Also, using the laser capture microdissection technique combined with RT-qPCR, we detected a substantial decrease in Pnoc but an increase in Oprl1 mRNA levels in the RMTg of Post-EtOH rats. Moreover, RMTg cFos expression is increased in Post-EtOH rats, which display anxiety- and depression-like behaviors. Intra-RMTg infusion of the endogenous NOP agonist nociceptin attenuates the aversive behaviors in Post-EtOH rats without causing any notable change in Naïve rats. Conversely, intra-RMTg infusion of the NOP selective antagonist [Nphe1]nociceptin(1-13)NH2 elicits anxiety- and depression-like behaviors in Naïve but not Post-EtOH rats. Furthermore, intra-RMTg infusion of nociceptin significantly reduces alcohol consumption. Thus, our results show that the deficiency of RMTg NOP signaling during alcohol withdrawal mediates anxiety- and depression-like behaviors. The intervention of NOP may help those individuals suffering from alcohol use disorders.
Collapse
Affiliation(s)
- Wenfu Li
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Zhiheng Ren
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Ying Tang
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yixin Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Shizhu Sun
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Ruxuan Ding
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jiawei Hou
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yunlin Mai
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Bo Zhan
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yingxin Zhu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Wanhong Zuo
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Rao Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
6
|
Yi T, Wang N, Huang J, Wang Y, Ren S, Hu Y, Xia J, Liao Y, Li X, Luo F, Ouyang Q, Li Y, Zheng Z, Xiao Q, Ren R, Yao Z, Tang X, Wang Y, Chen X, He C, Li H, Hu Z. A Sleep-Specific Midbrain Target for Sevoflurane Anesthesia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300189. [PMID: 36961096 PMCID: PMC10214273 DOI: 10.1002/advs.202300189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/02/2023] [Indexed: 05/27/2023]
Abstract
Sevoflurane has been the most widely used inhaled anesthetics with a favorable recovery profile; however, the precise mechanisms underlying its anesthetic action are still not completely understood. Here the authors show that sevoflurane activates a cluster of urocortin 1 (UCN1+ )/cocaine- and amphetamine-regulated transcript (CART+ ) neurons in the midbrain involved in its anesthesia. Furthermore, growth hormone secretagogue receptor (GHSR) is highly enriched in sevoflurane-activated UCN1+ /CART+ cells and is necessary for sleep induction. Blockade of GHSR abolishes the excitatory effect of sevoflurane on UCN1+ /CART+ neurons and attenuates its anesthetic effect. Collectively, their data suggest that anesthetic action of sevoflurane necessitates the GHSR activation in midbrain UCN1+ /CART+ neurons, which provides a novel target including the nucleus and receptor in the field of anesthesia.
Collapse
Affiliation(s)
- Tingting Yi
- Department of AnesthesiologySecond Affiliated HospitalThird Military Medical UniversityChongqing400037China
- Department of AnesthesiologyYongchuan HospitalChongqing Medical UniversityChongqing402160China
| | - Na Wang
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
- College of BioengineeringChongqing UniversityChongqing400044China
| | - Jing Huang
- Department of AnesthesiologySecond Affiliated HospitalThird Military Medical UniversityChongqing400037China
| | - Yaling Wang
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Shuancheng Ren
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Yiwen Hu
- Department of AnesthesiologySecond Affiliated HospitalThird Military Medical UniversityChongqing400037China
| | - Jianxia Xia
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Yixiang Liao
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Xin Li
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Fenlan Luo
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Qin Ouyang
- School of PharmacyThird Military Medical UniversityChongqing400038China
| | - Yu Li
- Department of AnesthesiologySecond Affiliated HospitalThird Military Medical UniversityChongqing400037China
| | - Ziyi Zheng
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Qin Xiao
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Rong Ren
- Sleep Medicine CenterDepartment of Respiratory and Critical Care MedicineMental Health CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Zhongxiang Yao
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Xiangdong Tang
- Sleep Medicine CenterDepartment of Respiratory and Critical Care MedicineMental Health CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Yanjiang Wang
- Department of NeurologyDaping HospitalThird Military Medical UniversityChongqing400042China
| | - Xiaowei Chen
- Brain Research CenterCollaborative Innovation Center for Brain ScienceThird Military Medical UniversityChongqing400038China
| | - Chao He
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
| | - Hong Li
- Department of AnesthesiologySecond Affiliated HospitalThird Military Medical UniversityChongqing400037China
| | - Zhian Hu
- Department of PhysiologyThird Military Medical UniversityChongqing400038China
- College of BioengineeringChongqing UniversityChongqing400044China
- Chongqing Institute for Brain and IntelligenceGuangyang Bay LaboratoryChongqing400064China
| |
Collapse
|
7
|
Reduction of stress responses in honey bees by synthetic ligands targeting an allatostatin receptor. Sci Rep 2022; 12:16760. [PMID: 36202961 PMCID: PMC9537510 DOI: 10.1038/s41598-022-20978-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 09/21/2022] [Indexed: 11/08/2022] Open
Abstract
Honey bees are of great economic and ecological importance, but are facing multiple stressors that can jeopardize their pollination efficiency and survival. Therefore, understanding the physiological bases of their stress response may help defining treatments to improve their resilience. We took an original approach to design molecules with this objective. We took advantage of the previous identified neuropeptide allatostatin A (ASTA) and its receptor (ASTA-R) as likely mediators of the honey bee response to a biologically relevant stressor, exposure to an alarm pheromone compound. A first series of ASTA-R ligands were identified through in silico screening using a homology 3D model of the receptor and in vitro binding experiments. One of these (A8) proved also efficient in vivo, as it could counteract two behavioral effects of pheromone exposure, albeit only in the millimolar range. This putative antagonist was used as a template for the chemical synthesis of a second generation of potential ligands. Among these, two compounds showed improved efficiency in vivo (in the micromolar range) as compared to A8 despite no major improvement in their affinity for the receptor in vitro. These new ligands are thus promising candidates for alleviating stress in honey bees.
Collapse
|
8
|
Ma Y, Giardino WJ. Neural circuit mechanisms of the cholecystokinin (CCK) neuropeptide system in addiction. ADDICTION NEUROSCIENCE 2022; 3:100024. [PMID: 35983578 PMCID: PMC9380858 DOI: 10.1016/j.addicn.2022.100024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Given historical focus on the roles for cholecystokinin (CCK) as a peripheral hormone controlling gastrointestinal processes and a brainstem peptide regulating food intake, the study of CCK as a limbic neuromodulator coordinating reward-seeking and emotional behavior remains underappreciated. Furthermore, localization of CCK to specialized interneurons throughout the hippocampus and cortex relegated CCK to being examined primarily as a static cell type marker rather than a dynamic functional neuromodulator. Yet, over three decades of literature have been generated by efforts to delineate the central mechanisms of addiction-related behaviors mediated by the CCK system across the striatum, amygdala, hypothalamus, and midbrain. Here, we cover fundamental findings that implicate CCK neuron activity and CCK receptor signaling in modulating drug intake and drug-seeking (focusing on psychostimulants, opioids, and alcohol). In doing so, we highlight the few studies that indicate sex differences in CCK expression and corresponding drug effects, emphasizing the importance of examining hormonal influences and sex as a biological variable in translating basic science discoveries to effective treatments for substance use disorders in human patients. Finally, we point toward understudied subcortical sources of endogenous CCK and describe how continued neurotechnology advancements can be leveraged to modernize understanding of the neural circuit mechanisms underlying CCK release and signaling in addiction-relevant behaviors.
Collapse
Affiliation(s)
- Yihe Ma
- Department of Psychiatry & Behavioral Sciences and Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - William J. Giardino
- Department of Psychiatry & Behavioral Sciences and Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
9
|
Domi A, Lunerti V, Petrella M, Domi E, Borruto AM, Ubaldi M, Weiss F, Ciccocioppo R. Genetic deletion or pharmacological blockade of nociceptin/orphanin FQ receptors in the ventral tegmental area attenuates nicotine-motivated behaviour. Br J Pharmacol 2022; 179:2647-2658. [PMID: 34854073 PMCID: PMC9081114 DOI: 10.1111/bph.15762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The nociceptin/orphanin FQ (N/OFQ)-nociceptin opioid-like peptide (NOP) receptor system is widely distributed in the brain and pharmacological activation of this system revealed therapeutic potential in animal models of substance use disorder. Studies also showed that genetic deletion or pharmacological blockade of NOP receptors confer resistance to the development of alcohol abuse. Here, we have used a genetic and pharmacological approach to evaluate the therapeutic potential of NOP antagonism in smoking cessation. EXPERIMENTAL APPROACH Constitutive NOP receptor knockout rats (NOP-/- ) and their wild-type counterparts (NOP+/+ ) were tested over a range of behaviours to characterize their motivation for nicotine. We next explored the effects of systemic administration of the NOP receptor antagonist LY2817412 (1.0 & 3.0 mg·kg-1 ) on nicotine self-administration. NOP receptor blockade was further evaluated at the brain circuitry level, by microinjecting LY2817412 (3.0 & 6.0 μg·μl-1 ) into the ventral tegmental area (VTA), nucleus accumbens (NAc) and central amygdala (CeA). KEY RESULTS Genetic NOP receptor deletion resulted in decreased nicotine intake, decreased motivation to self-administer and attenuation of cue-induced nicotine reinstatement. LY2817412 reduced nicotine intake in NOP+/+ but not in NOP-/- rats, confirming that its effect is mediated by inhibition of NOP transmission. Finally, injection of LY2817412 into the VTA but not into the NAc or CeA decreased nicotine self-administration. CONCLUSIONS AND IMPLICATIONS These findings indicate that inhibition of NOP transmission attenuates the motivation for nicotine through mechanisms involving the VTA and suggest that NOP receptor antagonism may represent a potential treatment for smoking cessation.
Collapse
Affiliation(s)
- Ana Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Veronica Lunerti
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Michele Petrella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Esi Domi
- Center for Social and Affective Neuroscience, Institute for Clinical and Experimental Medicine, Linkoping University, Linkoping 58183, Sweden
| | - Anna Maria Borruto
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
10
|
Discovery of a functionally selective ghrelin receptor (GHSR 1a) ligand for modulating brain dopamine. Proc Natl Acad Sci U S A 2022; 119:e2112397119. [PMID: 35239443 PMCID: PMC8915830 DOI: 10.1073/pnas.2112397119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The modulation of growth hormone secretagogue receptor-1a (GHSR1a) signaling is a promising strategy for treating brain conditions of metabolism, aging, and addiction. GHSR1a activation results in pleiotropic physiological outcomes through distinct and pharmacologically separable G protein– and β-arrestin (βarr)–dependent signaling pathways. Thus, pathway-selective modulation can enable improved pharmacotherapeutics that can promote therapeutic efficacy while mitigating side effects. Here, we describe the discovery of a brain-penetrant small molecule, N8279 (NCATS-SM8864), that biases GHSR1a conformations toward Gαq activation and reduces aberrant dopaminergic behavior in mice. N8279 represents a promising chemical scaffold to advance the development of better treatments for GHSR1a-related brain disorders involving the pathological dysregulation of dopamine. The growth hormone secretagogue receptor-1a (GHSR1a) is the cognate G protein–coupled receptor (GPCR) for the peptide hormone ghrelin. GHSR1a is a promising therapeutic target for a wide range of metabolic, age-related, and central nervous system (CNS)–based conditions. In addition, growing evidence supports that GHSR1a is a modulator of dopamine (DA) homeostasis and is neuroprotective within brain DA circuits. GHSR1a signaling originates from pharmacologically separable G protein– and β-arrestin (βarr)–dependent pathways, and consequently, GHSR1a-mediated physiological responses depend upon their distinctive signaling contributions. Thus, when treating disorders of disrupted DA homeostasis, a pharmacological strategy that modulates biased GHSR1a signaling may uncouple desired therapeutic outcomes from unwanted side effects. Here, we report the discovery of a small molecule GHSR1a agonist, N8279 (NCATS-SM8864), functionally selective for G protein signaling. Comprehensive pharmacological characterization reveals that N8279 elicits potent Gαq activity at the apo- and ghrelin-bound GHSR1a. Further biochemical analysis and molecular modeling demonstrate that N8279 signaling requires the extracellular domain of GHSR1a, especially extracellular loop 2. Collectively, these findings suggest that N8279 possesses an extended binding mode into the extracellular vestibule of the GHSR1a that preferentially favors Gαq signaling over alternative G proteins and βarr2-dependent cellular responses. Critically, N8279 is brain-penetrant in mice, exhibits CNS stability, and attenuates dysfunctional DA-mediated behaviors in both genetic and pharmacological mouse models of hyperdopaminergia. Our findings provide insight into the mechanisms governing GPCR functional selectivity and emphasize how biased ligand drug development can produce novel GHSR1a pharmacotherapeutics to treat pathological disruptions of brain DA homeostasis.
Collapse
|
11
|
Exploring the role of neuropeptides in depression and anxiety. Prog Neuropsychopharmacol Biol Psychiatry 2022; 114:110478. [PMID: 34801611 DOI: 10.1016/j.pnpbp.2021.110478] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/13/2021] [Accepted: 11/13/2021] [Indexed: 12/24/2022]
Abstract
Depression is one of the most prevalent forms of mental disorders and is the most common cause of disability in the Western world. Besides, the harmful effects of stress-related mood disorders on the patients themselves, they challenge the health care system with enormous social and economic impacts. Due to the high proportion of patients not responding to existing drugs, finding new treatment strategies has become an important topic in neurobiology, and there is much evidence that neuropeptides are not only involved in the physiology of stress but may also be clinically important. Based on preclinical trial data, new neuropharmaceutical candidates may target neuropeptides and their receptors and are expected to be essential and valuable tools in the treatment of psychiatric disorders. In the current article, we have summarized data obtained from animal models of depressive disorder and transgenic mouse models. We also focus on previously published research data of clinical studies on corticotropin-releasing hormone (CRH), galanin (GAL), neuropeptide Y (NPY), neuropeptide S (NPS), Oxytocin (OXT), vasopressin (VP), cholecystokinin (CCK), and melanin-concentrating hormone (MCH) stress research fields.
Collapse
|
12
|
Salim C, Kan AK, Batsaikhan E, Patterson EC, Jee C. Neuropeptidergic regulation of compulsive ethanol seeking in C. elegans. Sci Rep 2022; 12:1804. [PMID: 35110557 PMCID: PMC8810865 DOI: 10.1038/s41598-022-05256-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 01/07/2022] [Indexed: 11/09/2022] Open
Abstract
Despite the catastrophic consequences of alcohol abuse, alcohol use disorders (AUD) and comorbidities continue to strain the healthcare system, largely due to the effects of alcohol-seeking behavior. An improved understanding of the molecular basis of alcohol seeking will lead to enriched treatments for these disorders. Compulsive alcohol seeking is characterized by an imbalance between the superior drive to consume alcohol and the disruption or erosion in control of alcohol use. To model the development of compulsive engagement in alcohol seeking, we simultaneously exploited two distinct and conflicting Caenorhabditis elegans behavioral programs, ethanol preference and avoidance of aversive stimulus. We demonstrate that the C. elegans model recapitulated the pivotal features of compulsive alcohol seeking in mammals, specifically repeated attempts, endurance, and finally aversion-resistant alcohol seeking. We found that neuropeptide signaling via SEB-3, a CRF receptor-like GPCR, facilitates the development of ethanol preference and compels animals to seek ethanol compulsively. Furthermore, our functional genomic approach and behavioral elucidation suggest that the SEB-3 regulates another neuropeptidergic signaling, the neurokinin receptor orthologue TKR-1, to facilitate compulsive ethanol-seeking behavior.
Collapse
Affiliation(s)
- Chinnu Salim
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center (UTHSC), 71 S. Manassas St., Suite 217, Memphis, TN, 38103, USA
| | - Ann Ke Kan
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center (UTHSC), 71 S. Manassas St., Suite 217, Memphis, TN, 38103, USA
| | - Enkhzul Batsaikhan
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center (UTHSC), 71 S. Manassas St., Suite 217, Memphis, TN, 38103, USA
| | - E Clare Patterson
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center (UTHSC), 71 S. Manassas St., Suite 217, Memphis, TN, 38103, USA
| | - Changhoon Jee
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center (UTHSC), 71 S. Manassas St., Suite 217, Memphis, TN, 38103, USA.
| |
Collapse
|
13
|
Bhat US, Shahi N, Surendran S, Babu K. Neuropeptides and Behaviors: How Small Peptides Regulate Nervous System Function and Behavioral Outputs. Front Mol Neurosci 2021; 14:786471. [PMID: 34924955 PMCID: PMC8674661 DOI: 10.3389/fnmol.2021.786471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
One of the reasons that most multicellular animals survive and thrive is because of the adaptable and plastic nature of their nervous systems. For an organism to survive, it is essential for the animal to respond and adapt to environmental changes. This is achieved by sensing external cues and translating them into behaviors through changes in synaptic activity. The nervous system plays a crucial role in constantly evaluating environmental cues and allowing for behavioral plasticity in the organism. Multiple neurotransmitters and neuropeptides have been implicated as key players for integrating sensory information to produce the desired output. Because of its simple nervous system and well-established neuronal connectome, C. elegans acts as an excellent model to understand the mechanisms underlying behavioral plasticity. Here, we critically review how neuropeptides modulate a wide range of behaviors by allowing for changes in neuronal and synaptic signaling. This review will have a specific focus on feeding, mating, sleep, addiction, learning and locomotory behaviors in C. elegans. With a view to understand evolutionary relationships, we explore the functions and associated pathophysiology of C. elegans neuropeptides that are conserved across different phyla. Further, we discuss the mechanisms of neuropeptidergic signaling and how these signals are regulated in different behaviors. Finally, we attempt to provide insight into developing potential therapeutics for neuropeptide-related disorders.
Collapse
Affiliation(s)
- Umer Saleem Bhat
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Navneet Shahi
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Siju Surendran
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Kavita Babu
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
14
|
Ubaldi M, Cannella N, Borruto AM, Petrella M, Micioni Di Bonaventura MV, Soverchia L, Stopponi S, Weiss F, Cifani C, Ciccocioppo R. Role of Nociceptin/Orphanin FQ-NOP Receptor System in the Regulation of Stress-Related Disorders. Int J Mol Sci 2021; 22:12956. [PMID: 34884757 PMCID: PMC8657682 DOI: 10.3390/ijms222312956] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ) is a 17-residue neuropeptide that binds the nociceptin opioid-like receptor (NOP). N/OFQ exhibits nucleotidic and aminoacidics sequence homology with the precursors of other opioid neuropeptides but it does not activate either MOP, KOP or DOP receptors. Furthermore, opioid neuropeptides do not activate the NOP receptor. Generally, activation of N/OFQ system exerts anti-opioids effects, for instance toward opioid-induced reward and analgesia. The NOP receptor is widely expressed throughout the brain, whereas N/OFQ localization is confined to brain nuclei that are involved in stress response such as amygdala, BNST and hypothalamus. Decades of studies have delineated the biological role of this system demonstrating its involvement in significant physiological processes such as pain, learning and memory, anxiety, depression, feeding, drug and alcohol dependence. This review discusses the role of this peptidergic system in the modulation of stress and stress-associated psychiatric disorders in particular drug addiction, mood, anxiety and food-related associated-disorders. Emerging preclinical evidence suggests that both NOP agonists and antagonists may represent a effective therapeutic approaches for substances use disorder. Moreover, the current literature suggests that NOP antagonists can be useful to treat depression and feeding-related diseases, such as obesity and binge eating behavior, whereas the activation of NOP receptor by agonists could be a promising tool for anxiety.
Collapse
Affiliation(s)
- Massimo Ubaldi
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Nazzareno Cannella
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Anna Maria Borruto
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Michele Petrella
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Maria Vittoria Micioni Di Bonaventura
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Laura Soverchia
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Serena Stopponi
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - Carlo Cifani
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Roberto Ciccocioppo
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| |
Collapse
|
15
|
Pomrenze MB, Walker LC, Giardino WJ. Gray areas: Neuropeptide circuits linking the Edinger-Westphal and Dorsal Raphe nuclei in addiction. Neuropharmacology 2021; 198:108769. [PMID: 34481834 PMCID: PMC8484048 DOI: 10.1016/j.neuropharm.2021.108769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 01/16/2023]
Abstract
The circuitry of addiction comprises several neural networks including the midbrain - an expansive region critically involved in the control of motivated behaviors. Midbrain nuclei like the Edinger-Westphal (EW) and dorsal raphe (DR) contain unique populations of neurons that synthesize many understudied neuroactive molecules and are encircled by the periaqueductal gray (PAG). Despite the proximity of these special neuron classes to the ventral midbrain complex and surrounding PAG, functions of the EW and DR remain substantially underinvestigated by comparison. Spanning approximately -3.0 to -5.2 mm posterior from bregma in the mouse, these various cell groups form a continuum of neurons that we refer to collectively as the subaqueductal paramedian zone. Defining how these pathways modulate affective behavioral states presents a difficult, yet conquerable challenge for today's technological advances in neuroscience. In this review, we cover the known contributions of different neuronal subtypes of the subaqueductal paramedian zone. We catalogue these cell types based on their spatial, molecular, connectivity, and functional properties and integrate this information with the existing data on the EW and DR in addiction. We next discuss evidence that links the EW and DR anatomically and functionally, highlighting the potential contributions of an EW-DR circuit to addiction-related behaviors. Overall, we aim to derive an integrated framework that emphasizes the contributions of EW and DR nuclei to addictive states and describes how these cell groups function in individuals suffering from substance use disorders. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
Affiliation(s)
- Matthew B Pomrenze
- Dept. of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - William J Giardino
- Dept. of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA.
| |
Collapse
|
16
|
Tobinski AM, Rappeneau V. Role of the Neuropeptide S System in Emotionality, Stress Responsiveness and Addiction-Like Behaviours in Rodents: Relevance to Stress-Related Disorders. Pharmaceuticals (Basel) 2021; 14:ph14080780. [PMID: 34451877 PMCID: PMC8400992 DOI: 10.3390/ph14080780] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/22/2022] Open
Abstract
The neuropeptide S (NPS) and its receptor (NPSR1) have been extensively studied over the last two decades for their roles in locomotion, arousal/wakefulness and anxiety-related and fear-related behaviours in rodents. However, the possible implications of the NPS/NPSR1 system, especially those of the single nucleotide polymorphism (SNP) rs324981, in stress-related disorders and substance abuse in humans remain unclear. This is possibly due to the fact that preclinical and clinical research studies have remained separated, and a comprehensive description of the role of the NPS/NPSR1 system in stress-relevant and reward-relevant endpoints in humans and rodents is lacking. In this review, we describe the role of the NPS/NPSR1 system in emotionality, stress responsiveness and addiction-like behaviour in rodents. We also summarize the alterations in the NPS/NPSR1 system in individuals with stress-related disorders, as well as the impact of the SNP rs324981 on emotion, stress responses and neural activation in healthy individuals. Moreover, we discuss the therapeutic potential and possible caveats of targeting the NPS/NPSR1 system for the treatment of stress-related disorders. The primary goal of this review is to highlight the importance of studying some rodent behavioural readouts modulated by the NPS/NPSR1 system and relevant to stress-related disorders.
Collapse
|
17
|
Keller C, Wei P, Wancewicz B, Cross TWL, Rey FE, Li L. Extraction optimization for combined metabolomics, peptidomics, and proteomics analysis of gut microbiota samples. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4625. [PMID: 32885503 PMCID: PMC7855350 DOI: 10.1002/jms.4625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/28/2020] [Accepted: 07/11/2020] [Indexed: 05/02/2023]
Abstract
Multiomic studies are increasingly performed to gain a deeper understanding of molecular processes occurring in a biological system, such as the complex microbial communities (i.e., microbiota) that reside the distal gut. While a combination of metabolomics and proteomics is more commonly used, multiomics studies including peptidomcis characterization are less frequently undertaken. Here, we investigated three different extraction methods, chosen for their previous use in extracting metabolites, peptides, and proteins, and compared their ability to perform metabolomic, peptidomic, and proteomic analysis of mouse cecum content. The methanol/chloroform/water extraction performed the best for metabolomic and peptidomic analysis as it detected the largest number of small molecules and identified the largest number of peptides, but the acidified methanol extraction performed best for proteomics analysis as it had the highest number of protein identifications. The methanol/chloroform/water extraction was further analyzed by identifying metabolites with tandem mass spectrometry (MS/MS) analysis and by gene ontology analysis for the peptide and protein results to provide a multiomics analysis of the gut microbiota.
Collapse
Affiliation(s)
- Caitlin Keller
- Department of Chemistry, University of Wisconsin-Madison, Madison WI, 53705
| | - Pingli Wei
- Department of Chemistry, University of Wisconsin-Madison, Madison WI, 53705
| | - Benjamin Wancewicz
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison WI, 53705
| | - Tzu-Wen L Cross
- Department of Bacteriology, University of Wisconsin-Madison, Madison WI, 53705
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison WI, 53705
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison WI, 53705
- School of Pharmacy, University of Wisconsin-Madison, Madison WI, 53705
| |
Collapse
|
18
|
Fonville L, Paterson L, Herlinger K, Hayes A, Hill R, Nutt D, Lingford-Hughes A. Functional evaluation of NK 1 antagonism on cue reactivity in opiate dependence; An fMRI study. Drug Alcohol Depend 2021; 221:108564. [PMID: 33548897 PMCID: PMC8047866 DOI: 10.1016/j.drugalcdep.2021.108564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 10/26/2022]
Abstract
BACKGROUND Opiate addiction is a major health challenge with substantial societal cost. Though harm minimisation strategies have been effective, there is a growing need for new treatments for detoxification and relapse prevention. Preclinical research has found neurokinin 1 (NK1) receptors have prominent effects on opiate reward and reinforcement, and human studies have found NK1 antagonism led to reductions in craving and withdrawal. However, its effect on brain mechanisms in opiate addiction has not yet been examined. METHODS This study aims to assess the impact of NK1 antagonist aprepitant on heroin cue-elicited changes in blood-oxygenation level dependent (BOLD) signal in opiate dependent individuals undergoing detoxification. Participants will attend two scanning sessions and receive a single dose of aprepitant (320 mg) and a placebo in a randomised, cross-over design. During functional magnetic resonance imaging participants will undergo two runs of a cue reactivity task, which consists of passive viewing of drug cues or neutral cues in a block design fashion. We hypothesise that NK1 antagonism will attenuate the BOLD response to drug cues in the caudate nucleus and amygdala. Regions of interest were selected based on NK1 receptor density and their role in cue reactivity and craving.
Collapse
Affiliation(s)
- Leon Fonville
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, United Kingdom.
| | - Louise Paterson
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, United Kingdom
| | - Katherine Herlinger
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, United Kingdom
| | - Alexandra Hayes
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, United Kingdom
| | - Raymond Hill
- Department of Metabolism, Digestion and Reproduction, Imperial College London, United Kingdom
| | - David Nutt
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, United Kingdom
| | - Anne Lingford-Hughes
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, United Kingdom
| |
Collapse
|
19
|
Giardino WJ, Pomrenze MB. Extended Amygdala Neuropeptide Circuitry of Emotional Arousal: Waking Up on the Wrong Side of the Bed Nuclei of Stria Terminalis. Front Behav Neurosci 2021; 15:613025. [PMID: 33633549 PMCID: PMC7900561 DOI: 10.3389/fnbeh.2021.613025] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Sleep is fundamental to life, and poor sleep quality is linked to the suboptimal function of the neural circuits that process and respond to emotional stimuli. Wakefulness ("arousal") is chiefly regulated by circadian and homeostatic forces, but affective mood states also strongly impact the balance between sleep and wake. Considering the bidirectional relationships between sleep/wake changes and emotional dynamics, we use the term "emotional arousal" as a representative characteristic of the profound overlap between brain pathways that: (1) modulate wakefulness; (2) interpret emotional information; and (3) calibrate motivated behaviors. Interestingly, many emotional arousal circuits communicate using specialized signaling molecules called neuropeptides to broadly modify neural network activities. One major neuropeptide-enriched brain region that is critical for emotional processing and has been recently implicated in sleep regulation is the bed nuclei of stria terminalis (BNST), a core component of the extended amygdala (an anatomical term that also includes the central and medial amygdalae, nucleus accumbens shell, and transition zones betwixt). The BNST encompasses an astonishing diversity of cell types that differ across many features including spatial organization, molecular signature, biological sex and hormonal milieu, synaptic input, axonal output, neurophysiological communication mode, and functional role. Given this tremendous complexity, comprehensive elucidation of the BNST neuropeptide circuit mechanisms underlying emotional arousal presents an ambitious set of challenges. In this review, we describe how rigorous investigation of these unresolved questions may reveal key insights to enhancing psychiatric treatments and global psychological wellbeing.
Collapse
|
20
|
Romero-Leguizamón CR, Kohlmeier KA. Stress-related endogenous neuropeptides induce neuronal excitation in the Laterodorsal Tegmentum. Eur Neuropsychopharmacol 2020; 38:86-97. [PMID: 32768153 DOI: 10.1016/j.euroneuro.2020.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/29/2020] [Accepted: 07/16/2020] [Indexed: 01/07/2023]
Abstract
Stress is a physiological response that promotes maintenance of balance against harmful stimuli. Unfortunately, chronic activation of stress systems facilitates the development of psychiatric disorders. A stress-mediated hypercholinergic state could underlie this facilitation, as cholinergic mechanisms have been suggested to play a role in anxiety, depression, and substance use disorder (SUD). Stimulation by stress hormones, urocortin (Ucn1) or corticotropin-releasing factor (CRF), of the CRF receptor type 1 (CRFR1) of acetylcholine-containing neurons of the laterodorsal tegmental nucleus (LDT) could be involved in modulation of cholinergic transmission during periods of stress hormone activation, which could play a role in psychiatric disorders as cholinergic LDT neurons project to, and control activity in, mood-, arousal- and SUD-controlling regions. The present study investigated for the first time the membrane effects and intracellular outcomes of CRFR1 activation by endogenous stress hormones on LDT neurons. Patch clamp recordings of immunohistochemically-identified cholinergic and non-cholinergic LDT neurons with concurrent calcium imaging were used to monitor cellular responses to CRFR1 stimulation with Ucn1 and CRF. Postsynaptically-mediated excitatory currents were elicited in LDT cholinergic neurons, accompanied by an enhancement in synaptic events. In addition, CRFR1 activation resulted in rises in intracellular calcium levels. CRFR1 stimulation recruited MAPK/ERK and SERCA-ATPase involved pathways. The data presented here provide the first evidence that Ucn1 and CRF exert pre and postsynaptic excitatory membrane actions on LDT cholinergic neurons that could underlie the hypercholinergic state associated with stress which could play a role in the heightened risk of psychiatric disorders associated with a chronic stress state.
Collapse
Affiliation(s)
- Cesar R Romero-Leguizamón
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, 2100, Denmark.
| |
Collapse
|
21
|
Abstract
Addiction to substances such as alcohol, cocaine, opioids, and methamphetamine poses a continuing clinical and public challenge globally. Despite progress in understanding substance use disorders, challenges remain in their treatment. Some of these challenges include limited ability of therapeutics to reach the brain (blood-brain barrier), adverse systemic side effects of current medications, and importantly key aspects of addiction not addressed by currently available treatments (such as cognitive impairment). Inability to sustain abstinence or seek treatment due to cognitive deficits such as poor decision-making and impulsivity is known to cause poor treatment outcomes. In this review, we provide an evidenced-based rationale for intranasal drug delivery as a viable and safe treatment modality to bypass the blood-brain barrier and target insulin to the brain to improve the treatment of addiction. Intranasal insulin with improvement of brain cell energy and glucose metabolism, stress hormone reduction, and improved monoamine transmission may be an ideal approach for treating multiple domains of addiction including memory and impulsivity. This may provide additional benefits to enhance current treatment approaches.
Collapse
Affiliation(s)
- Bhavani Kashyap
- HealthPartners Neuroscience Center, 295 Phalen Blvd, St Paul, Minnesota, 55130, USA.
- HealthPartners Institute, Bloomington, Minnesota, USA.
| | - Leah R Hanson
- HealthPartners Neuroscience Center, 295 Phalen Blvd, St Paul, Minnesota, 55130, USA
- HealthPartners Institute, Bloomington, Minnesota, USA
| | - William H Frey Ii
- HealthPartners Neuroscience Center, 295 Phalen Blvd, St Paul, Minnesota, 55130, USA
- HealthPartners Institute, Bloomington, Minnesota, USA
| |
Collapse
|
22
|
Jordan CJ, Cao J, Newman AH, Xi ZX. Progress in agonist therapy for substance use disorders: Lessons learned from methadone and buprenorphine. Neuropharmacology 2019; 158:107609. [PMID: 31009632 PMCID: PMC6745247 DOI: 10.1016/j.neuropharm.2019.04.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/25/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
Substance use disorders (SUD) are serious public health problems worldwide. Although significant progress has been made in understanding the neurobiology of drug reward and the transition to addiction, effective pharmacotherapies for SUD remain limited and a majority of drug users relapse even after a period of treatment. The United States Food and Drug Administration (FDA) has approved several medications for opioid, nicotine, and alcohol use disorders, whereas none are approved for the treatment of cocaine or other psychostimulant use disorders. The medications approved by the FDA for the treatment of SUD can be divided into two major classes - agonist replacement therapies, such as methadone and buprenorphine for opioid use disorders (OUD), nicotine replacement therapy (NRT) and varenicline for nicotine use disorders (NUD), and antagonist therapies, such as naloxone for opioid overdose and naltrexone for promoting abstinence. In the present review, we primarily focus on the pharmacological rationale of agonist replacement strategies in treatment of opioid dependence, and the potential translation of this rationale to new therapies for cocaine use disorders. We begin by describing the neural mechanisms underlying opioid reward, followed by preclinical and clinical findings supporting the utility of agonist therapies in the treatment of OUD. We then discuss recent progress of agonist therapies for cocaine use disorders based on lessons learned from methadone and buprenorphine. We contend that future studies should identify agonist pharmacotherapies that can facilitate abstinence in patients who are motivated to quit their illicit drug use. Focusing on those that are able to achieve abstinence from cocaine will provide a platform to broaden the effectiveness of medication and psychosocial treatment strategies for this underserved population. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.
Collapse
Affiliation(s)
- Chloe J Jordan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|
23
|
Witkiewitz K, Litten RZ, Leggio L. Advances in the science and treatment of alcohol use disorder. SCIENCE ADVANCES 2019; 5:eaax4043. [PMID: 31579824 PMCID: PMC6760932 DOI: 10.1126/sciadv.aax4043] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/28/2019] [Indexed: 05/03/2023]
Abstract
Alcohol is a major contributor to global disease and a leading cause of preventable death, causing approximately 88,000 deaths annually in the United States alone. Alcohol use disorder is one of the most common psychiatric disorders, with nearly one-third of U.S. adults experiencing alcohol use disorder at some point during their lives. Alcohol use disorder also has economic consequences, costing the United States at least $249 billion annually. Current pharmaceutical and behavioral treatments may assist patients in reducing alcohol use or facilitating alcohol abstinence. Although recent research has expanded understanding of alcohol use disorder, more research is needed to identify the neurobiological, genetic and epigenetic, psychological, social, and environmental factors most critical in the etiology and treatment of this disease. Implementation of this knowledge in clinical practice and training of health care providers is also needed to ensure appropriate diagnosis and treatment of individuals suffering from alcohol use disorder.
Collapse
Affiliation(s)
- K. Witkiewitz
- Department of Psychology and Center on Alcoholism, Substance Abuse, and Addictions, University of New Mexico, 2650 Yale Blvd. SE, Albuquerque, NM 87106, USA
| | - R. Z. Litten
- Division of Medications Development and Division of Treatment and Recovery Research, National Institute on Alcohol Abuse and Alcoholism, 6700B Rockledge Drive, Bethesda, MD 20892-6902, USA
| | - L. Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, and National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 10 Center Drive (10CRC/15330), Bethesda, MD 21224, USA
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd., Baltimore, MD 21224, USA
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI 02912, USA
- Corresponding author.
| |
Collapse
|
24
|
Sliedrecht W, de Waart R, Witkiewitz K, Roozen HG. Alcohol use disorder relapse factors: A systematic review. Psychiatry Res 2019; 278:97-115. [PMID: 31174033 DOI: 10.1016/j.psychres.2019.05.038] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022]
Abstract
A relapsing-remitting course is very common in patients with an Alcohol Use Disorder (AUD). Understanding the determinants associated with alcohol resumption remains a formidable task. This paper examines relapse determinants based on a systematic review of recent alcohol literature (2000-2019). Relevant databases were consulted for articles that contained information about specific relapse determinants and reported statistical significance of each relapse determinant in predicting relapse. Relapse was broadly defined based on the characterization in the included articles. From the initial identified 4613 papers, a total of 321 articles were included. Results encompass multiple relapse determinants, which were ordered according to biopsychosocial and spiritual categories, and presented, using a descriptive methodology. Psychiatric co-morbidity, AUD severity, craving, use of other substances, health and social factors were consistently significantly associated with AUD relapse. Conversely, supportive social network factors, self efficacy, and factors related to purpose and meaning in life, were protective against AUD relapse. Despite heterogeneity in different methods, measures, and sample characteristics, these findings may contribute to a better therapeutic understanding in which specific factors are associated with relapse and those that prevent relapse. Such factors may have a role in a personalized medicine framework to improve patient outcomes.
Collapse
Affiliation(s)
- Wilco Sliedrecht
- De Hoop GGZ, Provincialeweg 70, 3329 KP Dordrecht, the Netherlands.
| | - Ranne de Waart
- Mentrum/Arkin, Wisselwerking 46-48, 1112 XR Diemen, the Netherlands.
| | - Katie Witkiewitz
- The University of New Mexico (UNM), MSC 03-2220, Univ of New Mexico, Albuquerque, NM 87131, USA.
| | - Hendrik G Roozen
- The University of New Mexico (UNM), Center on Alcoholism, Substance Abuse, and Addictions (CASAA), MSC 11 6280, 1 Univ of New Mexico, Albuquerque, NM 87106, USA.
| |
Collapse
|
25
|
Escalated Alcohol Self-Administration and Sensitivity to Yohimbine-Induced Reinstatement in Alcohol Preferring Rats: Potential Role of Neurokinin-1 Receptors in the Amygdala. Neuroscience 2019; 413:77-85. [PMID: 31242442 DOI: 10.1016/j.neuroscience.2019.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/08/2019] [Accepted: 06/15/2019] [Indexed: 12/16/2022]
Abstract
Genetic factors significantly contribute to the risk for developing alcoholism. To study these factors and other associated phenotypes, rodent lines have been developed using selective breeding for high alcohol preference. One of these models, the alcohol preferring (P) rat, has been used in hundreds of preclinical studies over the last few decades. However, very few studies have examined relapse-like behavior in this rat strain. In this study, we used operant self-administration and yohimbine-induced reinstatement models to examine relapse-like behavior in P rats. Our previous work has demonstrated that P rats show increased expression of the neurokinin-1 receptor (NK1R) in the central nucleus of the amygdala (CeA), and this functionally contributes to escalated alcohol consumption in this strain. We hypothesized that P rats would show increased sensitivity to yohimbine-induced reinstatement that is also mediated by NK1R in the CeA. Using Fos staining, site-specific infusion of NK1R antagonist, and viral vector overexpression, we examined the influence of NK1R on the sensitivity to yohimbine-induced reinstatement of alcohol seeking. We found that P rats displayed increased sensitivity to yohimbine-induced reinstatement as well as increased neuronal activation in the CeA after yohimbine injection compared to the control Wistar strain. Intra-CeA infusion of NK1R antagonist attenuates yohimbine-induced reinstatement in P rats. Conversely, upregulation of NK1R within the CeA of Wistar rats increases alcohol consumption and sensitivity to yohimbine-induced reinstatement. These findings suggest that NK1R upregulation in the CeA contributes to multiple alcohol-related phenotypes in the P rat, including alcohol consumption and sensitivity to relapse.
Collapse
|
26
|
Terenina EE, Cavigelli S, Mormede P, Zhao W, Parks C, Lu L, Jones BC, Mulligan MK. Genetic Factors Mediate the Impact of Chronic Stress and Subsequent Response to Novel Acute Stress. Front Neurosci 2019; 13:438. [PMID: 31164799 PMCID: PMC6536627 DOI: 10.3389/fnins.2019.00438] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/16/2019] [Indexed: 12/31/2022] Open
Abstract
Individual differences in physiological and biobehavioral adaptation to chronic stress are important predictors of health and fitness; genetic differences play an important role in this adaptation. To identify these differences we measured the biometric, neuroendocrine, and transcriptional response to stress among inbred mouse strains with varying degrees of genetic similarity, C57BL/6J (B), C57BL/6NJ (N), and DBA/2J (D). The B and D strains are highly genetically diverse whereas the B and N substrains are highly similar. Strain differences in hypothalamic-pituitary-adrenal (HPA) axis cross-sensitization were determined by plasma corticosterone (CORT) levels and hippocampal gene expression following 7-weeks of chronic mild stress (CMS) or normal housing (NH) and subsequent exposure to novel acute restraint. Fecal CORT metabolites and body and organ weights were also measured. All strains exposed to CMS had reduced heart weights, whereas body weight gain was attenuated only in B and N strains. Acute stress alone produced larger plasma CORT responses in the D and N strains compared to the B strain. CMS paired with acute stress produced cross-sensitization of the CORT response in the N strain. The N strain also had the largest number of hippocampal transcripts with up-regulated expression in response to stress. In contrast, the D strain had the largest number of transcripts with down-regulated expression following CMS and acute stress. In summary, we observed differential responses to CMS at both the physiological and molecular level among genetically diverse strains, indicating that genetic factors drive individual differences in experience-dependent regulation of the stress response.
Collapse
Affiliation(s)
- Elena E Terenina
- GenPhySE, ENVT, INRA, Université de Toulouse, Castanet-Tolosan, France.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sonia Cavigelli
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Pierre Mormede
- GenPhySE, ENVT, INRA, Université de Toulouse, Castanet-Tolosan, France.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wenyuan Zhao
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Cory Parks
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Byron C Jones
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
27
|
Chowdhury VS. Heat Stress Biomarker Amino Acids and Neuropeptide Afford Thermotolerance in Chicks. J Poult Sci 2019; 56:1-11. [PMID: 32055190 PMCID: PMC6993887 DOI: 10.2141/jpsa.0180024] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/16/2018] [Indexed: 12/18/2022] Open
Abstract
With global warming, heat stress is becoming a pressing concern worldwide. In chickens, heat stress reduces food intake and growth, and increases body temperature and stress responses. Although it is believed that young chicks do not experience heat stress as they need a higher ambient temperature to survive, our series of studies in young chicks showed that they are sensitive to heat stress. This review summarizes current knowledge on amino acid metabolisms during heat stress, with special emphasis on the hypothermic functions of l-citrulline (l-Cit) and l-leucine (l-Leu), and the functions of neuropeptide Y (NPY) in terms of body temperature and heat stress regulation in chicks. Amino acid metabolism is severely affected by heat stress. For example, prolonged heat stress reduces plasma l-Cit in chicks and l-Leu in the brain and liver of embryos. l-Cit and l-Leu supplementation affords thermotolerance in young chicks. NPY expression is increased in the brains of heat-exposed chicks. NPY has a hypothermic action under control thermoneutral temperature and heat stress in chicks. The NPY-sub-receptor Y5 is a partial mediator of the hypothermic action of NPY. Further, NPY stimulates brain dopamine concentrations and acts as an anti-stress agent in heat-exposed fasted, but not fed chicks. In conclusion, young chicks can serve as a model animal for the study of heat stress in chickens. l-Cit, l-Leu, and NPY were identified as biomarkers of heat stress, with the potential to afford thermotolerance in chicks.
Collapse
Affiliation(s)
- Vishwajit S. Chowdhury
- Lab of Stress Physiology and Metabolism, Graduate School of Bioresource and Bioenvironmental Science, Division for Experimental Natural Science, Faculty of Arts and Science, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
28
|
Ehlers CL, Gilder DA, Gizer IR, Wilhelmsen KC. Indexing the 'dark side of addiction': substance-induced affective symptoms and alcohol use disorders. Addiction 2019; 114:139-149. [PMID: 30153346 PMCID: PMC6320236 DOI: 10.1111/add.14431] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/13/2018] [Accepted: 08/20/2018] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND AIMS The emergence of negative affective symptoms during the course of alcohol use disorders (AUDs) (e.g. 'dark side' symptoms) has been suggested theoretically; however, the description of their occurrence is limited. This study operationalized two negative affect symptoms and tested the strength of association between these phenotypes and (1) indicators of the clinical course of the severity of AUD, (2) comorbid Axis I psychiatric disorders, suicidal behaviors and trait neuroticism and (3) whether participants reported drinking to relieve the negative affective symptoms. DESIGN A retrospective cross-sectional study was used to evaluate associations, using logistic regression, between the two negative affective symptoms and clinical measures of AUD severity and progression as well as comorbidity with other psychiatric disorders and conditions, adjusted for demographic characteristics. SETTING US community-based studies. PARTICIPANTS A total of 2568 individuals with AUDs obtained from larger population studies that targeted individuals of European American (n = 1663), Mexican American and American Indian (n = 905) ancestry. MEASUREMENTS Semi-Structured Assessment for the Genetics of Alcoholism was used to ascertain the two 'dark side' phenotypes, clinical diagnoses, the clinical course of AUD and associated symptoms. The two phenotypes were: (1) being anxious or depressed when trying to cut down or stop drinking and (2) experiencing disabling depression for more than 24 hours while drinking. FINDINGS Both phenotypes were found to be rare in mild and moderate use disorder and highly prevalent in severe AUDs. Having an independent anxiety or affective disorder and elevated scores on trait neuroticism were also associated significantly with the occurrence of both symptoms, as was alcohol 'craving', elevated treatment-seeking, suicidal behaviors and drinking to relieve the symptoms. CONCLUSIONS Affective symptoms are common in severe alcohol use disorders are associated with a history of independent affective/anxiety disorders, neuroticism and suicidal behaviors; and may promote further heavy drinking.
Collapse
Affiliation(s)
- Cindy L Ehlers
- Department of Neurosciences, The Scripps Research Institute, La Jolla, CA, USA
| | - David A Gilder
- Department of Neurosciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Kirk C Wilhelmsen
- Departments of Genetics and Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
29
|
Micioni Di Bonaventura MV, Micioni Di Bonaventura E, Cifani C, Polidori C. N/OFQ-NOP System in Food Intake. Handb Exp Pharmacol 2019; 254:279-295. [PMID: 31073870 DOI: 10.1007/164_2019_212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
While lifestyle modifications should be the first-line actions in preventing and treating obesity and eating disorders, pharmacotherapy also provides a necessary tool for the management of these diseases.However, given the limitations of current anti-obesity drugs, innovative treatments that improve efficacy and safety are needed.Since the discovery that the activation of the Nociceptin/Orphanin (N/OFQ) FQ peptide (NOP) receptor by N/OFQ induces an increase of food intake in laboratory animals, and the finding that this effect can be blocked by NOP antagonists, many NOP agonists and antagonists have been synthesized and tested in vitro and in vivo for their potential regulation of feeding behavior. Promising results seem to suggest that the N/OFQergic system may be a potential therapeutic target for the neural control of feeding behavior and related pathologies, especially in binge-like eating behavior.
Collapse
Affiliation(s)
| | | | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, MC, Italy.
| | - Carlo Polidori
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, MC, Italy
| |
Collapse
|
30
|
Yang N, Anapindi KDB, Romanova EV, Rubakhin SS, Sweedler JV. Improved identification and quantitation of mature endogenous peptides in the rodent hypothalamus using a rapid conductive sample heating system. Analyst 2018; 142:4476-4485. [PMID: 29098220 DOI: 10.1039/c7an01358b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Measurement, identification, and quantitation of endogenous peptides in tissue samples by mass spectrometry (MS) contribute to our understanding of the complex molecular mechanisms of numerous biological phenomena. For accurate results, it is essential to arrest the postmortem degradation of ubiquitous proteins in samples prior to performing peptidomic measurements. Doing so ensures that the detection of endogenous peptides, typically present at relatively low levels of abundance, is not overwhelmed by protein degradation products. Heat stabilization has been shown to inactivate the enzymes in tissue samples and minimize the presence of protein degradation products in the subsequent peptide extracts. However, the efficacy of different heat treatments to preserve the integrity of full-length endogenous peptides has not been well documented; prior peptidomic studies of heat stabilization methods have not distinguished between the full-length (mature) and numerous truncated (possible artifacts of sampling) forms of endogenous peptides. We show that thermal sample treatment via rapid conductive heat transfer is effective for detection of mature endogenous peptides in fresh and frozen rodent brain tissues. Freshly isolated tissue processing with the commercial Stabilizor T1 heat stabilization system resulted in the confident identification of 65% more full-length mature neuropeptides compared to widely used sample treatment in a hot water bath. This finding was validated by a follow-up quantitative multiple reaction monitoring MS analysis of select neuropeptides. The rapid conductive heating in partial vacuum provided by the Stabilizor T1 effectively reduces protein degradation and decreases the chemical complexity of the sample, as assessed by determining total protein content. This system enabled the detection, identification, and quantitation of neuropeptides related to 22 prohormones expressed in individual rat hypothalami and suprachiasmatic nuclei.
Collapse
Affiliation(s)
- Ning Yang
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana 61801, USA.
| | | | | | | | | |
Collapse
|
31
|
Xu Y, Jia J, Xie C, Wu Y, Tu W. Transient Receptor Potential Ankyrin 1 and Substance P Mediate the Development of Gastric Mucosal Lesions in a Water Immersion Restraint Stress Rat Model. Digestion 2018; 97:228-239. [PMID: 29428952 DOI: 10.1159/000484980] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/04/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND Activation of substance P (SP) contributes to the development and maintenance of gastric lesions, but the mechanisms underlying the release of SP and SP-mediated damage to the gastric mucosa remain unknown. Transient receptor potential ankyrin 1 (TRPA1) is expressed in SP-positive neurons in the dorsal root ganglion (DRG) and stomach of rats. We hypothesized that water immersion restraint stress (WIRS) may activate and sensitize TRPA1 in DRG neurons, subsequently inducing the release of SP from DRG and stomach cells, causing the development of acute gastric mucosal lesions (AGML). METHODS Changes in TRPA1 and SP expression in T8-11 DRG sensory neurons and the stomach in an AGML rat model were determined by reverse transcription polymerase chain reaction, western blotting and immunohistochemistry. The SP levels of serum and gastric mucosa were measured by using an enzyme-linked immunosorbent assay (ELISA). Gastric lesions were evaluated by histopathological changes. The TRPA1 antagonist HC-030031 and TRPA1 agonists allyl isothiocyanate were used to verify effect of TRPA1 and SP on AGML. RESULTS SP and TRPA1 in the DRG and stomach were upregulated, and the serum and gastric mucosa levels of SP were increased after WIRS, which are closely associated with AGML. The release of SP was suppressed and AGML were alleviated following a selective TRPA1 antagonist HC-030031. TRPA1 agonists AITC increased release of SP and led to moderate gastric lesions. We confirmed that WIRS induced the release of SP in the DRG, stomach, serum and gastric mucosa, and in a TRPA1-dependent manner. CONCLUSIONS Upregulated SP and TRPA1 in the DRG and stomach and increased serum and gastric mucosa SP levels may contribute to stress-induced AGML. TRPA1 is a potential drug target to reduce stress-induced AGML development in patients with acute critical illnesses. This study may contribute to the discovery of drugs for AGML treatment.
Collapse
Affiliation(s)
- Yan Xu
- Department of anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, The Second Military Medical University, Guangzhou, China.,Department of Anesthesiology, the 173rd Clinical Department of PLA, 421rd Hospital, Huizhou, China
| | - Ji Jia
- Department of anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, The Second Military Medical University, Guangzhou, China
| | - Chuangbo Xie
- Department of anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, The Second Military Medical University, Guangzhou, China
| | - Youping Wu
- Department of anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, The Second Military Medical University, Guangzhou, China
| | - Weifeng Tu
- Department of anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, The Second Military Medical University, Guangzhou, China
| |
Collapse
|
32
|
Thiel D, Franz-Wachtel M, Aguilera F, Hejnol A. Xenacoelomorph Neuropeptidomes Reveal a Major Expansion of Neuropeptide Systems during Early Bilaterian Evolution. Mol Biol Evol 2018. [PMCID: PMC6188537 DOI: 10.1093/molbev/msy160] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuropeptides are neurosecretory signaling molecules in protostomes and deuterostomes (together Nephrozoa). Little, however, is known about the neuropeptide complement of the sister group of Nephrozoa, the Xenacoelomorpha, which together form the Bilateria. Because members of the xenacoelomorph clades Xenoturbella, Nemertodermatida, and Acoela differ extensively in their central nervous system anatomy, the reconstruction of the xenacoelomorph and bilaterian neuropeptide complements may provide insights into the relationship between nervous system evolution and peptidergic signaling. Here, we analyzed transcriptomes of seven acoels, four nemertodermatids, and two Xenoturbella species using motif searches, similarity searches, mass spectrometry and phylogenetic analyses to characterize neuropeptide precursors and neuropeptide receptors. Our comparison of these repertoires with previously reported nephrozoan and cnidarian sequences shows that the majority of annotated neuropeptide GPCRs in cnidarians are not orthologs of specific bilaterian neuropeptide receptors, which suggests that most of the bilaterian neuropeptide systems evolved after the cnidarian–bilaterian evolutionary split. This expansion of more than 20 peptidergic systems in the stem leading to the Bilateria predates the evolution of complex nephrozoan organs and nervous system architectures. From this ancient set of neuropeptides, acoels show frequent losses that correlate with their divergent central nervous system anatomy. We furthermore detected the emergence of novel neuropeptides in xenacoelomorphs and their expansion along the nemertodermatid and acoel lineages, the two clades that evolved nervous system condensations. Together, our study provides fundamental insights into the early evolution of the bilaterian peptidergic systems, which will guide future functional and comparative studies of bilaterian nervous systems.
Collapse
Affiliation(s)
- Daniel Thiel
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway
| | | | - Felipe Aguilera
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway
| | - Andreas Hejnol
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway
| |
Collapse
|
33
|
The neurokinin-1 receptor mediates escalated alcohol intake induced by multiple drinking models. Neuropharmacology 2018; 137:194-201. [PMID: 29758386 DOI: 10.1016/j.neuropharm.2018.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/16/2022]
Abstract
We have previously demonstrated that the neurokinin-1 receptor (NK1R) is upregulated in the central nucleus of the amygdala of alcohol preferring (P) rats and that this receptor mediates escalated alcohol consumption in this strain. However, it is unclear if non-genetic models of escalated consumption are also mediated by NK1R signaling, and if so, what brain regions govern this effect. In the experiments presented here, we use two methods of inducing escalated alcohol intake in outbred Wistar rats: yohimbine pretreatment and intermittent alcohol access (Monday, Wednesday, and Friday availability; 20% alcohol). We found that escalated alcohol consumption induced by both yohimbine injection and intermittent access is attenuated by systemic administration of the NK1R antagonist L822429. Also, when compared to continuous alcohol access or access to water alone, NK1R expression was increased in the nucleus accumbens (NAC) and dorsal striatum, but not the amygdala. Escalated consumption induced by intermittent access was attenuated when the NK1R antagonist L822429 was infused directly into the dorsal striatum, but not when infused into the NAC. Taken together, these results suggest that NK1R upregulation contributes to escalated alcohol consumption that is induced by genetic selection, yohimbine injection, and intermittent access. However there is a dissociation between the regions involved in these behaviors with amygdalar upregulation contributing to genetic predisposition to escalated consumption and striatal upregulation driving escalation that is induced by environmental exposures.
Collapse
|
34
|
Li W, Papilloud A, Lozano-Montes L, Zhao N, Ye X, Zhang X, Sandi C, Rainer G. Stress Impacts the Regulation Neuropeptides in the Rat Hippocampus and Prefrontal Cortex. Proteomics 2018; 18:e1700408. [PMID: 29406625 DOI: 10.1002/pmic.201700408] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/10/2018] [Indexed: 11/05/2022]
Abstract
Adverse life experiences increase the lifetime risk to several stress-related psychopathologies, such as anxiety or depressive-like symptoms following stress in adulthood. However, the neurochemical modulations triggered by stress have not been fully characterized. Neuropeptides play an important role as signaling molecules that contribute to physiological regulation and have been linked to neurological and psychiatric diseases. However, little is known about the influence of stress on neuropeptide regulation in the brain. Here, we have performed an exploratory study of how neuropeptide expression at adulthood is modulated by experiencing a period of multiple stressful experiences. We have targeted hippocampus and prefrontal cortex (PFC) brain areas, which have previously been shown to be modulated by stressors, employing a targeted liquid chromatography-mass spectrometry (LC-MS) based approach that permits broad peptide coverage with high sensitivity. We found that in the hippocampus, Met-enkephalin, Met-enkephalin-Arg-Phe, and Met-enkephalin-Arg-Gly-Leu were upregulated, while Leu-enkephalin and Little SAAS were downregulated after stress. In the PFC area, Met-enkephalin-Arg-Phe, Met-enkephalin-Arg-Gly-Leu, peptide PHI-27, somatostatin-28 (AA1-12), and Little SAAS were all downregulated. This systematic evaluation of neuropeptide alterations in the hippocampus and PFC suggests that stressors impact neuropeptides and that neuropeptide regulation is brain-area specific. These findings suggest several potential peptide candidates, which warrant further investigations in terms of correlation with depression-associated behaviors.
Collapse
Affiliation(s)
- Wenxue Li
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Aurelie Papilloud
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Science, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | | | - Nan Zhao
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Xueting Ye
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Xiaozhe Zhang
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Science, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Gregor Rainer
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
35
|
Kubrak OI, Lushchak OV, Zandawala M, Nässel DR. Systemic corazonin signalling modulates stress responses and metabolism in Drosophila. Open Biol 2017; 6:rsob.160152. [PMID: 27810969 PMCID: PMC5133436 DOI: 10.1098/rsob.160152] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/05/2016] [Indexed: 12/20/2022] Open
Abstract
Stress triggers cellular and systemic reactions in organisms to restore homeostasis. For instance, metabolic stress, experienced during starvation, elicits a hormonal response that reallocates resources to enable food search and readjustment of physiology. Mammalian gonadotropin-releasing hormone (GnRH) and its insect orthologue, adipokinetic hormone (AKH), are known for their roles in modulating stress-related behaviour. Here we show that corazonin (Crz), a peptide homologous to AKH/GnRH, also alters stress physiology in Drosophila. The Crz receptor (CrzR) is expressed in salivary glands and adipocytes of the liver-like fat body, and CrzR knockdown targeted simultaneously to both these tissues increases the fly's resistance to starvation, desiccation and oxidative stress, reduces feeding, alters expression of transcripts of Drosophila insulin-like peptides (DILPs), and affects gene expression in the fat body. Furthermore, in starved flies, CrzR-knockdown increases circulating and stored carbohydrates. Thus, our findings indicate that elevated systemic Crz signalling during stress coordinates increased food intake and diminished energy stores to regain metabolic homeostasis. Our study suggests that an ancient stress-peptide in Urbilateria evolved to give rise to present-day GnRH, AKH and Crz signalling systems.
Collapse
Affiliation(s)
- Olga I Kubrak
- Department of Zoology, Stockholm University, 10691 Stockholm, Sweden
| | - Oleh V Lushchak
- Department of Zoology, Stockholm University, 10691 Stockholm, Sweden
| | - Meet Zandawala
- Department of Zoology, Stockholm University, 10691 Stockholm, Sweden
| | - Dick R Nässel
- Department of Zoology, Stockholm University, 10691 Stockholm, Sweden
| |
Collapse
|
36
|
Kaye JT, Bradford DE, Magruder KP, Curtin JJ. Probing for Neuroadaptations to Unpredictable Stressors in Addiction: Translational Methods and Emerging Evidence. J Stud Alcohol Drugs 2017; 78:353-371. [PMID: 28499100 DOI: 10.15288/jsad.2017.78.353] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stressors clearly contribute to addiction etiology and relapse in humans, but our understanding of specific mechanisms remains limited. Rodent models of addiction offer the power, flexibility, and precision necessary to delineate the causal role and specific mechanisms through which stressors influence alcohol and other drug use. This review describes a program of research using startle potentiation to unpredictable stressors that is well positioned to translate between animal models and clinical research with humans on stress neuroadaptations in addiction. This research rests on a solid foundation provided by three separate pillars of evidence from (a) rodent behavioral neuroscience on stress neuroadaptations in addiction, (b) rodent affective neuroscience on startle potentiation, and (c) human addiction and affective science with startle potentiation. Rodent stress neuroadaptation models implicate adaptations in corticotropin-releasing factor and norepinephrine circuits within the central extended amygdala following chronic alcohol and other drug use that mediate anxious behaviors and stress-induced reinstatement among drug-dependent rodents. Basic affective neuroscience indicates that these same neural mechanisms are involved in startle potentiation to unpredictable stressors in particular (vs. predictable stressors). We believe that synthesis of these evidence bases should focus us on the role of unpredictable stressors in addiction etiology and relapse. Startle potentiation in unpredictable stressor tasks is proposed to provide an attractive and flexible test bed to encourage tight translation and reverse translation between animal models and human clinical research on stress neuroadaptations. Experimental therapeutics approaches focused on unpredictable stressors hold high promise to identify, repurpose, or refine pharmacological and psychosocial interventions for addiction.
Collapse
Affiliation(s)
- Jesse T Kaye
- University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | | |
Collapse
|
37
|
Di Bonaventura MVM, Ubaldi M, Giusepponi ME, Rice KC, Massi M, Ciccocioppo R, Cifani C. Hypothalamic CRF1 receptor mechanisms are not sufficient to account for binge-like palatable food consumption in female rats. Int J Eat Disord 2017; 50:1194-1204. [PMID: 28833350 PMCID: PMC5772704 DOI: 10.1002/eat.22767] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 07/29/2017] [Accepted: 08/01/2017] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The present study evaluated the effect of systemic injection of the CRF1 receptor antagonist R121919, the corticosterone synthesis inhibitor metyrapone and central amygdala (CeA) injections of the nonselective CRF antagonist D-Phe-CRF(12-41) in rats in which binge eating was evoked by stress and cycles of food restriction. METHOD Female rats were subjected or not to repeated cycles of regular chow food restriction/ad libitum feeding during which they were also given limited access (2 h) to palatable food. On the test day, rats were either exposed or not to the sight of the palatable food for 15 min without allowing access, before assessing food consumption. RESULTS Systemic injections of R121919, but not of the metyrapone, blocked binge-like eating behavior. Restricted and stressed rats showed up-regulation of crh1 receptor mRNA signal in the bed nucleus of the stria terminalis and CeA but not in basolateral amygdala (BLA) or in the paraventricular nucleus. Injection D-Phe-CRF(12-41) in CeA but not in the BLA-blocked binge-like eating behavior. DISCUSSION These findings demonstrate that extra-hypothalamic CRF1 receptors, rather than those involved in endocrine functions, are involved in binge eating and the crucial role of CRF receptors in CeA. CRF1 receptor antagonism may represent a novel pharmacological treatment for binge-related eating disorders.
Collapse
Affiliation(s)
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | | | - Kenner C. Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse (NIDA) and National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892-3373, United States
| | - Maurizio Massi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy,NIDA/NIH, Intramural Research Program, 21224 Baltimore (MD), USA
| |
Collapse
|
38
|
Reduction of smoking urges with intranasal insulin: a randomized, crossover, placebo-controlled clinical trial. Mol Psychiatry 2017; 22:1413-1421. [PMID: 28242873 DOI: 10.1038/mp.2016.234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/20/2016] [Accepted: 10/31/2016] [Indexed: 11/09/2022]
Abstract
Many cigarette smokers express a desire to quit smoking, but ~85% of cessation attempts fail. In our attempt to delineate genetic modulators of smoking persistence, we have earlier shown that a locus within an ~250 kb haplotype block spanning the 5' untranslated region region of insulin-degrading enzyme is associated with serum cotinine levels; the study's measure of smoking quantity. Based on our findings, and coupled with recent preclinical studies showing the importance of multiple neuropeptides in reinstatement of drug use, we formulated intranasal insulin to evaluate its efficacy during acute abstinence from smoking. Our original study was a crossover trial including 19 otherwise healthy smokers who abstained from smoking for 36 h. The morning following their second night of abstinence, in random order, study participants received intranasal insulin (60 IU) or placebo (8.7% sodium chloride). The goal of our second study was to replicate the craving findings from the original trial and expand this research by including additional stress-related measures. Thirty-seven study participants abstained from smoking overnight. The next day, they were administered either intranasal insulin (60 IU) or placebo, following which they participated in the Trier Social Stress Test Task. This was a parallel design study focusing on the standard stress subjective, hormonal and cardiovascular measures. We also evaluated any changes in circulating glucose, insulin and c-peptide (a marker of endogenous insulin). In the original study, intranasal insulin significantly reduced morning nicotine craving (b=3.65, P⩽0.05). Similarly, in the second study, intranasal insulin reduced nicotine cravings over time (b=0.065, P⩽0.05) and the effect lasted through the psychosocial stress period. Intranasal insulin also increased circulating cortisol levels (F=12.78, P⩽0.001). No changes in insulin or c-peptide were detected. A significant treatment × time interaction (P⩽0.05) was detected for glucose, but subjects remained well within the euglycemic range. Previous studies have shown that heightened nicotine cravings and blunted response to stress are independent and significant predictors of relapse to smoking. In our study, intranasal insulin normalized the subjective and hormonal response to stress. As such, intranasal insulin should further be studied in a larger clinical trial of smoking cessation. In support of this, we provide evidence that the treatment is safe and effective and, based on absence of peripheral insulin changes, conclude that the pharmacodynamic effect is centrally driven.
Collapse
|
39
|
Ye H, Wang J, Tian Z, Ma F, Dowell J, Bremer Q, Lu G, Baldo B, Li L. Quantitative Mass Spectrometry Reveals Food Intake-Induced Neuropeptide Level Changes in Rat Brain: Functional Assessment of Selected Neuropeptides as Feeding Regulators. Mol Cell Proteomics 2017; 16:1922-1937. [PMID: 28864778 DOI: 10.1074/mcp.ra117.000057] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Indexed: 12/27/2022] Open
Abstract
Endogenous neuropeptides are important signaling molecules that function as regulators of food intake and body weight. Previous work has shown that neuropeptide gene expression levels in a forebrain reward site, the nucleus accumbens (NAc), were changed by feeding. To directly monitor feeding-induced changes in neuropeptide expression levels within the NAc, we employed a combination of cryostat dissection, heat stabilization, neuropeptide extraction and label-free quantitative neuropeptidomics via a liquid chromatography-high resolution mass spectrometry platform. Using this methodology, we described the first neuropeptidome in NAc and discovered that feeding caused the expression level changes of multiple neuropeptides derived from different precursors, especially proSAAS-derived peptides such as Big LEN, PEN and little SAAS. We further investigated the regulatory functions of these neuropeptides derived from the ProSAAS family by performing an intra-NAc microinjection experiment using the identified ProSAAS neuropeptides, 'Big-LEN' and 'PEN'. Big LEN significantly increased rats' food and water intake, whereas both big LEN and PEN affected other behaviors including locomotion, drinking and grooming. In addition, we quantified the feeding-induced changes of peptides from hippocampus, hypothalamus and striatum to reveal the neuropeptide interplay among different anatomical regions. In summary, our study demonstrated neuropeptidomic changes in response to food intake in the rat NAc and other key brain regions. Importantly, the microinfusion of ProSAAS peptides into NAc revealed that they are behaviorally active in this brain site, suggesting the potential use of these peptides as therapeutics for eating disorders.
Collapse
Affiliation(s)
- Hui Ye
- From the ‡State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Tongjiaxiang #24, Nanjing 21009, China.,§School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53705
| | - Jingxin Wang
- ¶Neuroscience Training Program, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705
| | - Zichuan Tian
- ‖Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706
| | - Fengfei Ma
- §School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53705
| | - James Dowell
- §School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53705
| | - Quentin Bremer
- **Department of Psychiatry, University of Wisconsin-Madison, 6001 Research Park Boulevard, Madison, Wisconsin 53719
| | - Gaoyuan Lu
- From the ‡State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Tongjiaxiang #24, Nanjing 21009, China
| | - Brian Baldo
- ¶Neuroscience Training Program, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705; .,**Department of Psychiatry, University of Wisconsin-Madison, 6001 Research Park Boulevard, Madison, Wisconsin 53719
| | - Lingjun Li
- §School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53705; .,¶Neuroscience Training Program, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705.,‖Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706.,‡‡School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
40
|
Nennig SE, Schank JR. The Role of NFkB in Drug Addiction: Beyond Inflammation. Alcohol Alcohol 2017; 52:172-179. [PMID: 28043969 DOI: 10.1093/alcalc/agw098] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
Aims Nuclear factor kappa light chain enhancer of activated B cells (NFkB) is a ubiquitous transcription factor well known for its role in the innate immune response. As such, NFkB is a transcriptional activator of inflammatory mediators such as cytokines. It has recently been demonstrated that alcohol and other drugs of abuse can induce NFkB activity and cytokine expression in the brain. A number of reviews have been published highlighting this effect of alcohol, and have linked increased NFkB function to neuroimmune-stimulated toxicity. However, in this review we focus on the potentially non-immune functions of NFkB as possible links between NFkB and addiction. Methods An extensive review of the literature via Pubmed searches was used to assess the current state of the field. Results NFkB can induce the expression of a diverse set of gene targets besides inflammatory mediators, some of which are involved in addictive processes, such as opioid receptors and neuropeptides. NFkB mediates complex behaviors including learning and memory, stress responses, anhedonia and drug reward, processes that may lie outside the role of NFkB in the classic neuroimmune response. Conclusions Future studies should focus on these non-immune functions of NFkB signaling and their association with addiction-related processes.
Collapse
Affiliation(s)
- S E Nennig
- Department of Physiology and Pharmacology, University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602, USA
| | - J R Schank
- Department of Physiology and Pharmacology, University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602, USA
| |
Collapse
|
41
|
Contribution of Urocortin to the Development of Excessive Drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:275-291. [PMID: 29056154 DOI: 10.1016/bs.irn.2017.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The corticotropin-releasing factor (CRF) system plays a role in alcohol consumption, and its dysregulation can contribute to alcohol use disorder. This system includes four peptide ligands: CRF, urocortin (Ucn)1, Ucn2, and Ucn3. Historically, attention focused on CRF, however, Ucn1 also plays a critical role in excessive alcohol use. This review covers evidence for this contribution and contrasts the role of Ucn1 with CRF. While CRF can promote binge consumption, this regulation occurs through generalized mechanisms that are not specific for alcohol. In contrast, inhibition of Ucn1 action specifically blunts escalation of alcohol drinking. Lesions, genetic knockout, and RNA interference experiments indicate that the centrally projecting Edinger-Westphal nucleus is the neuroanatomical source of Ucn1 critical for alcohol drinking. We propose that the contributions of Ucn1 to excessive drinking likely occur through enhancing rewarding properties of alcohol and symptoms of alcohol withdrawal, whereas CRF drives dependence-induced drinking at later stages of alcohol use. The transition from occasional binge drinking to dependence intricately depends on CRF system plasticity and coordination of CRF and Ucn1.
Collapse
|
42
|
Becker HC. Influence of stress associated with chronic alcohol exposure on drinking. Neuropharmacology 2017; 122:115-126. [PMID: 28431971 PMCID: PMC5497303 DOI: 10.1016/j.neuropharm.2017.04.028] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/12/2017] [Accepted: 04/17/2017] [Indexed: 12/24/2022]
Abstract
Stress is commonly regarded as an important trigger for relapse and a significant factor that promotes increased motivation to drink in some individuals. However, the relationship between stress and alcohol is complex, likely changing in form during the transition from early moderated alcohol use to more heavy uncontrolled alcohol intake. A growing body of evidence indicates that prolonged excessive alcohol consumption serves as a potent stressor, producing persistent dysregulation of brain reward and stress systems beyond normal homeostatic limits. This progressive dysfunctional (allostatic) state is characterized by changes in neuroendocrine and brain stress pathways that underlie expression of withdrawal symptoms that reflect a negative affective state (dysphoria, anxiety), as well as increased motivation to self-administer alcohol. This review highlights literature supportive of this theoretical framework for alcohol addiction. In particular, evidence for stress-related neural, physiological, and behavioral changes associated with chronic alcohol exposure and withdrawal experience is presented. Additionally, this review focuses on the effects of chronic alcohol-induced changes in several pro-stress neuropeptides (corticotropin-releasing factor, dynorphin) and anti-stress neuropeptide systems (nocicepton, neuropeptide Y, oxytocin) in contributing to the stress, negative emotional, and motivational consequences of chronic alcohol exposure. Studies involving use of animal models have significantly increased our understanding of the dynamic stress-related physiological mechanisms and psychological underpinnings of alcohol addiction. This, in turn, is crucial for developing new and more effective therapeutics for treating excessive, harmful drinking, particularly stress-enhanced alcohol consumption. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Department of Neuroscience, Medical University of South Carolina, RHJ Department of Veterans Affairs, Charleston, SC 29464, USA.
| |
Collapse
|
43
|
Anderson RI, Becker HC. Role of the Dynorphin/Kappa Opioid Receptor System in the Motivational Effects of Ethanol. Alcohol Clin Exp Res 2017; 41:1402-1418. [PMID: 28425121 DOI: 10.1111/acer.13406] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022]
Abstract
Evidence has demonstrated that dynorphin (DYN) and the kappa opioid receptor (KOR) system contribute to various psychiatric disorders, including anxiety, depression, and addiction. More recently, this endogenous opioid system has received increased attention as a potential therapeutic target for treating alcohol use disorders. In this review, we provide an overview and synthesis of preclinical studies examining the influence of alcohol (ethanol [EtOH]) exposure on DYN/KOR expression and function, as well as studies examining the effects of DYN/KOR manipulation on EtOH's rewarding and aversive properties. We then describe work that has characterized effects of KOR activation and blockade on EtOH self-administration and EtOH dependence/withdrawal-related behaviors. Finally, we address how the DYN/KOR system may contribute to stress-EtOH interactions. Despite an apparent role for the DYN/KOR system in motivational effects of EtOH, support comes from relatively few studies. Nevertheless, review of this literature reveals several common themes: (i) rodent strains genetically predisposed to consume more EtOH generally appear to have reduced DYN/KOR tone in brain reward circuitry; (ii) acute and chronic EtOH exposure typically up-regulate the DYN/KOR system; (iii) KOR antagonists reduce behavioral indices of negative affect associated with stress and chronic EtOH exposure/withdrawal; and (iv) KOR antagonists are effective in reducing EtOH consumption, but are often more efficacious under conditions that engender high levels of consumption, such as dependence or stress exposure. These results support the contention that the DYN/KOR system plays a significant role in contributing to dependence- and stress-induced elevation in EtOH consumption. Overall, more comprehensive analyses (on both behavioral and mechanistic levels) are needed to provide additional insight into how the DYN/KOR system is engaged and adapts to influence the motivation effects of EtOH. This information will be critical for the development of new pharmacological agents targeting KORs as promising novel therapeutics for alcohol use disorders and comorbid affective disorders.
Collapse
Affiliation(s)
- Rachel I Anderson
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina.,Department of Neuroscience , Medical University of South Carolina, Charleston, South Carolina.,RHJ Department of Veterans Affairs Medical Center , Charleston, South Carolina
| |
Collapse
|
44
|
Compton P, Chang YP. Substance Abuse and Addiction: Implications for Pain Management in Patients With Cancer. Clin J Oncol Nurs 2017; 21:203-209. [DOI: 10.1188/17.cjon.203-209] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
45
|
Schank JR, Heilig M. Substance P and the Neurokinin-1 Receptor: The New CRF. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:151-175. [DOI: 10.1016/bs.irn.2017.06.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
46
|
Giardino WJ, Rodriguez ED, Smith ML, Ford MM, Galili D, Mitchell SH, Chen A, Ryabinin AE. Control of chronic excessive alcohol drinking by genetic manipulation of the Edinger-Westphal nucleus urocortin-1 neuropeptide system. Transl Psychiatry 2017; 7:e1021. [PMID: 28140406 PMCID: PMC5299395 DOI: 10.1038/tp.2016.293] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/25/2016] [Accepted: 12/15/2016] [Indexed: 11/23/2022] Open
Abstract
Midbrain neurons of the centrally projecting Edinger-Westphal nucleus (EWcp) are activated by alcohol, and enriched with stress-responsive neuropeptide modulators (including the paralog of corticotropin-releasing factor, urocortin-1). Evidence suggests that EWcp neurons promote behavioral processes for alcohol-seeking and consumption, but a definitive role for these cells remains elusive. Here we combined targeted viral manipulations and gene array profiling of EWcp neurons with mass behavioral phenotyping in C57BL/6 J mice to directly define the links between EWcp-specific urocortin-1 expression and voluntary binge alcohol intake, demonstrating a specific importance for EWcp urocortin-1 activity in escalation of alcohol intake.
Collapse
Affiliation(s)
- W J Giardino
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - E D Rodriguez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - M L Smith
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - M M Ford
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - D Galili
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - S H Mitchell
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - A Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA,Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA. E-mail:
| |
Collapse
|
47
|
Gilpin NW, Weiner JL. Neurobiology of comorbid post-traumatic stress disorder and alcohol-use disorder. GENES BRAIN AND BEHAVIOR 2016; 16:15-43. [PMID: 27749004 DOI: 10.1111/gbb.12349] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/03/2016] [Accepted: 10/07/2016] [Indexed: 12/12/2022]
Abstract
Post-traumatic stress disorder (PTSD) and alcohol-use disorder (AUD) are highly comorbid in humans. Although we have some understanding of the structural and functional brain changes that define each of these disorders, and how those changes contribute to the behavioral symptoms that define them, little is known about the neurobiology of comorbid PTSD and AUD, which may be due in part to a scarcity of adequate animal models for examining this research question. The goal of this review is to summarize the current state-of-the-science on comorbid PTSD and AUD. We summarize epidemiological data documenting the prevalence of this comorbidity, review what is known about the potential neurobiological basis for the frequent co-occurrence of PTSD and AUD and discuss successes and failures of past and current treatment strategies. We also review animal models that aim to examine comorbid PTSD and AUD, highlighting where the models parallel the human condition, and we discuss the strengths and weaknesses of each model. We conclude by discussing key gaps in our knowledge and strategies for addressing them: in particular, we (1) highlight the need for better animal models of the comorbid condition and better clinical trial design, (2) emphasize the need for examination of subpopulation effects and individual differences and (3) urge cross-talk between basic and clinical researchers that is reflected in collaborative work with forward and reverse translational impact.
Collapse
Affiliation(s)
- N W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA.,Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - J L Weiner
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
48
|
Pucci M, Micioni Di Bonaventura MV, Giusepponi ME, Romano A, Filaferro M, Maccarrone M, Ciccocioppo R, Cifani C, D'Addario C. Epigenetic regulation of nociceptin/orphanin FQ and corticotropin-releasing factor system genes in frustration stress-induced binge-like palatable food consumption. Addict Biol 2016; 21:1168-1185. [PMID: 26387568 DOI: 10.1111/adb.12303] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 08/03/2015] [Accepted: 08/11/2015] [Indexed: 01/17/2023]
Abstract
Evidence suggests that binge eating may be caused by a unique interaction between dieting and stress. We developed a binge-eating model in which female rats with a history of intermittent food restriction show binge-like palatable food consumption after a 15-minute exposure to the sight of the palatable food (frustration stress). The aim of the present study was to investigate the regulation of the stress neurohormone corticotropin-releasing factor (CRF) system and of the nociceptin/orphanin FQ (N/OFQ) system genes in selective rat brain regions, using our animal model. Food restriction by itself seems to be responsible in the hypothalamus for the downregulation on messenger RNA levels of CRF-1 receptor, N/OFQ and its receptor (NOP). For the latter, this alteration might be due to selective histone modification changes. Instead, CRF gene appears to be upregulated in the hypothalamus as well as in the ventral tegmental area only when rats are food restricted and exposed to frustration stress, and, of relevance, these changes appear to be due to a reduction in DNA methylation at gene promoters. Moreover, also CRF-1 receptor gene resulted to be differentially regulated in these two brain regions. Epigenetic changes may be viewed as adaptive mechanisms to environmental perturbations concurring to facilitate food consumption in adverse conditions, that is, in this study, under food restriction and stressful conditions. Our data on N/OFQ and CRF signaling provide insight on the use of this binge-eating model for the study of epigenetic modifications in controlled genetic and environmental backgrounds.
Collapse
Affiliation(s)
- Mariangela Pucci
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Italy
| | | | | | - Adele Romano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Italy
| | - Monica Filaferro
- Department of Biomedical, Metabolic Sciences and Neurosciences, University of Modena and Reggio Emilia, Italy
| | - Mauro Maccarrone
- Campus Bio-Medico University of Rome, Italy
- European Center for Brain Research (CERC)/Santa Lucia Foundation, Italy
| | | | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy.
- Intramural Research Program, NIDA/NIH, Baltimore,, MD, USA.
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Italy.
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Sweden.
| |
Collapse
|
49
|
Ellenbroek BA, Angelucci F, Husum H, Mathé AA. Gene-environment interactions in a rat model of depression. Maternal separation affects neurotensin in selected brain regions. Neuropeptides 2016; 59:83-88. [PMID: 27372546 DOI: 10.1016/j.npep.2016.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/13/2016] [Accepted: 05/23/2016] [Indexed: 12/11/2022]
Abstract
Although the etiology of major psychiatric disorders has not been elucidated, accumulating evidence indicates that both genetic and early environmental factors play a role. We have previously demonstrated behavioral and neurochemical changes both in non-manipulated genetic rat models of depression, such as Flinders Sensitive Line (FSL) and Fawn Hooded (FH), and in normal rats following maternal separation (MS). The aim of the present study was to extend this work by exploring whether neurotensin (NT), a peptide implicated in several psychiatric disorders, is altered in a new animal model based on gene - environment interactions. More specifically, we used the FSL rats as a genetic model of depression and the Flinders Resistant Line (FRL) as controls and subjected them to MS. Pups randomly assigned to the MS procedure were separated from the dam as a litter for 180min daily between postnatal day 2 to 14. On postnatal day 90, rats were weighed and sacrificed by a two second high energy focused microwave irradiation and several brain regions were obtained by micropuncture. Neurotensin-like immunoreactivity (NT-LI) was measured by radioimmunoassay (RIA). The results showed that the FSL rats compared to the FRL rats have higher baseline NT-LI concentrations in the temporal cortex and periaqueductal gray and a markedly different response to maternal separation. The only observed change following maternal separation in the FRL rats was an NT-LI increase in the periaqueductal gray. In contrast, in the FSL significant increases were found in the nucleus accumbens, hippocampus, and entorhinal cortex and a decrease was seen in the temporal cortex after MS. The present study revealed baseline regional differences in NT-LI concentrations between the FSL and FRL strains and demonstrated that early MD differentially affects the two strains. The relevance of these alterations for depression as well as possible mechanisms underlying this gene-environment interaction are discussed.
Collapse
Affiliation(s)
- Bart A Ellenbroek
- School of Psychology, Victoria University of Wellington, Wellington, New Zealand.
| | | | - Henriette Husum
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Aleksander A Mathé
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Carboni L, Romoli B, Romualdi P, Zoli M. Repeated nicotine exposure modulates prodynorphin and pronociceptin levels in the reward pathway. Drug Alcohol Depend 2016; 166:150-8. [PMID: 27430399 DOI: 10.1016/j.drugalcdep.2016.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Nicotine dependence is maintained by neurobiological adaptations in the dopaminergic brain reward pathway with the contribution of opioidergic circuits. This study assessed the role of opioid peptides and receptors on the molecular changes associated with nicotine dependence. To this aim we analysed nicotine effects on opioid gene and receptor expression in the reward pathway in a nicotine sensitization model. METHODS Sprague-Dawley rats received nicotine administrations for five days and locomotor activity assessment showed the development of sensitization. The mRNA expression of prodynorphin (pdyn), pronociceptin (pnoc) and the respective receptors was measured by quantitative PCR in the ventral midbrain (VM), the nucleus accumbens (NAc), the caudate-putamen (CPu), the pre-frontal cortex (PFCx), and the hippocampus. RESULTS A significant positive effect of sensitization on pdyn mRNA levels was detected in the CPu. This effect was supported by a significant and selective correlation between the two parameters in this region. Moreover, chronic but not acute nicotine treatment significantly decreased pdyn mRNA levels in the NAc and increased expression in the PFCx. Pnoc mRNA was significantly increased in the VM and the PFCx after sub-chronic administration of nicotine, whereas no alterations were observed after acute treatment. No treatment associated changes were detected in κ-opioid receptor or nociceptin receptor mRNAs. CONCLUSIONS This experiment revealed an effect of nicotine administration that was distinguishable from the effect of nicotine sensitization. While several pnoc and pdyn changes were associated to nicotine administration, the only significant effect of sensitization was a significant increase in pdyn in the CPu.
Collapse
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy.
| | - Benedetto Romoli
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|