51
|
Xu L, Wang J. Food restriction in adolescence increases emotional disorder-like behaviors in adult rats. J Chem Neuroanat 2019; 104:101731. [PMID: 31862402 DOI: 10.1016/j.jchemneu.2019.101731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/14/2019] [Accepted: 12/15/2019] [Indexed: 10/25/2022]
Abstract
This study was designed to investigate the neuronal mechanism underlying the influence of early-life food restriction on the central nervous system and subsequent behaviors in adult rats. Several behavioral paradigms were tested in rats, including sucrose negative contrast test, forced swimming test (FST) and elevated plus maze test (EPM). in vivo intracellular electrophysiological recordings were conducted in the lateral nucleus of the amygdala (LAT). Finally, the levels of neuropeptide Y (NPY) were examined using immunohistochemistry. Food restriction during adolescence reduced sucrose preference in adult rats. Adolescent food restriction increased total immobile time in the FST and reduced the latency in rats to the first bout of immobility. In the EPM test, rats that experienced food restriction in adolescence and tested four weeks later spent less time than unrestricted controls in the open arm. In addition, chronic food restriction in adolescence increased in vivo LAT neuronal excitability in adulthood. Finally, NPY immunoreactivity in the LAT was reduced in rats that experienced chronic food restriction in adolescence compared to controls. Our results suggest that food restriction in adolescence increases emotional disorder-like behaviors in adult life, in which NPY production regulates the LAT-dependent behaviors and may underly the vulnerability to emotional disorders.
Collapse
Affiliation(s)
- Li Xu
- Department of Electrophysiological Examination, Cangzhou Central Hospital, Cangzhou 061001, Hebei Province, China
| | - Jinfeng Wang
- Department of Electrophysiological Examination, Cangzhou Central Hospital, Cangzhou 061001, Hebei Province, China.
| |
Collapse
|
52
|
Wyrofsky RR, Reyes BAS, Yu D, Kirby LG, Van Bockstaele EJ. Sex differences in the effect of cannabinoid type 1 receptor deletion on locus coeruleus-norepinephrine neurons and corticotropin releasing factor-mediated responses. Eur J Neurosci 2019; 48:2118-2138. [PMID: 30103253 DOI: 10.1111/ejn.14103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/06/2018] [Accepted: 08/07/2018] [Indexed: 11/30/2022]
Abstract
Cannabinoids are capable of modulating mood, arousal, cognition and behavior, in part via their effects on the noradrenergic nucleus locus coeruleus (LC). Dysregulation of LC signaling and norepinephrine (NE) efflux in the medial prefrontal cortex (mPFC) can lead to the development of psychiatric disorders, and CB1r deletion results in alterations of α2- and β1-adrenoceptors in the mPFC, suggestive of increased LC activity. To determine how CB1r deletion alters LC signaling, whole-cell patch-clamp electrophysiology was conducted in LC-NE neurons of male and female wild type (WT) and CB1r-knock out (KO) mice. CB1r deletion caused a significant increase in LC-NE excitability and input resistance in male but not female mice when compared to WT. CB1r deletion also caused adaptations in several indices of noradrenergic function. CB1r/CB2r-KO male mice had a significant increase in cortical NE levels and tyrosine hydroxylase and CRF levels in the LC compared to WT males. CB1r/CB2r-KO female mice showed a significant increase in LC α2-AR levels compared to WT females. To further probe actions of the endocannabinoid system as an anti-stress neuromediator, the effect of CB1r deletion on CRF-induced responses in the LC was investigated. The increase in LC-NE excitability observed in male and female WT mice following CRF (300 nM) bath application was not observed in CB1r-KO mice. These results indicate that cellular adaptations following CB1r deletion cause a disruption in LC-NE signaling in males but not females, suggesting underlying sex differences in compensatory mechanisms in KO mice as well as basal endocannabinoid regulation of LC-NE activity.
Collapse
Affiliation(s)
- Ryan R Wyrofsky
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, Pennsylvania
| | - Beverly A S Reyes
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, Pennsylvania
| | - Daohai Yu
- Department of Clinical Sciences, Temple Clinical Research Institute, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Lynn G Kirby
- Department of Anatomy and Cell Biology, Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Elisabeth J Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
53
|
Lee B, Yeom M, Shim I, Lee H, Hahm DH. Umbelliferone modulates depression-like symptoms by altering monoamines in a rat post-traumatic stress disorder model. J Nat Med 2019; 74:377-386. [DOI: 10.1007/s11418-019-01373-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/13/2019] [Indexed: 12/23/2022]
|
54
|
Yang Y, Yu H, Babygirija R, Shi B, Sun W, Zheng X, Zheng J. Intranasal Administration of Oxytocin Attenuates Stress Responses Following Chronic Complicated Stress in Rats. J Neurogastroenterol Motil 2019; 25:611-622. [PMID: 31587552 PMCID: PMC6786441 DOI: 10.5056/jnm19065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/04/2019] [Accepted: 07/20/2019] [Indexed: 12/13/2022] Open
Abstract
Background/Aims Gastrointestinal (GI) symptoms may develop when we fail to adapt to various stressors of our daily life. Central oxytocin (OXT) can counteract the biological actions of corticotropin-releasing factor (CRF), and in turn attenuates stress responses. Administration (intracerebroventricular) of OXT significantly antagonized the inhibitory effects of chronic complicated stress (CCS) on GI dysmotility in rats. However, intracerebroventricular administration is an invasive pathway. Intranasal administration can rapidly deliver peptides to the brain avoiding stress response. The effects of intranasal OXT on hypothalamus-pituitary-adrenal axis and GI motility in CCS conditions have not been investigated. Methods A CCS rat model was set up, OXT 5, 10, or 20 μg were intranasal administered, 30 minutes prior to stress loading. Central CRF and OXT expression levels were analyzed, serum corticosterone and OXT concentrations were measured, and gastric and colonic motor functions were evaluated by gastric emptying, fecal pellet output, and motility recording system. Results Rats in CCS condition showed significantly increased CRF expression and corticosterone concentration, which resulted in delayed gastric emptying and increased fecal pellet output, attenuated gastric motility and enhanced colonic motility were also recorded. OXT 10 μg or 20 μg significantly reduced CRF mRNA expression and the corticosterone concentration, OXT 20 μg also helped to restore GI motor dysfunction induced by CCS. Conclusion Intranasal administration of OXT has an anxiolytic effect and attenuates the hypothalamus-pituitary-adrenal axis in response to CCS, and gave effects which helped to restore GI dysmotility, and might be a new approach for the treatment of stress-induced GI motility disorders.
Collapse
Affiliation(s)
- Yu Yang
- Department of Physiology, School of Life Science, China Medical University, Shenyang, Liaoning Province, China
| | - Haijie Yu
- Department of Cardiology, First Affiliated Hospital, China Medical University, Shenyang, Liaoning Province, China
| | - Reji Babygirija
- Department of Surgery, Medical College of Wisconsin and Zablocki VA Medical Center, Milwaukee, WI, USA
| | - Bei Shi
- Department of Physiology, School of Life Science, China Medical University, Shenyang, Liaoning Province, China
| | - Weinan Sun
- Department of Physiology, School of Life Science, China Medical University, Shenyang, Liaoning Province, China
| | - Xiaojiao Zheng
- Department of Physiology, School of Life Science, China Medical University, Shenyang, Liaoning Province, China
| | - Jun Zheng
- Department of Physiology, School of Life Science, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
55
|
Maymon N, Mizrachi Zer-Aviv T, Sabban EL, Akirav I. Neuropeptide Y and cannabinoids interaction in the amygdala after exposure to shock and reminders model of PTSD. Neuropharmacology 2019; 162:107804. [PMID: 31622603 DOI: 10.1016/j.neuropharm.2019.107804] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 12/14/2022]
Abstract
Modulation of cannabinoid and neuropeptide Y (NPY) receptors may offer therapeutic benefits for post-traumatic stress disorder (PTSD). In this study, we aimed to investigate the functional interaction between these systems in the basolateral amygdala (BLA) in a rat model of PTSD. Rats were exposed to the shock and reminders model of PTSD and tested for hyper arousal/PTSD- and depression-like behaviors 3 weeks later. Immediately after shock exposure rats were microinjected into the BLA with URB597, a selective inhibitor of fatty acid amide hydrolase (FAAH) that increases the levels of the endocannabinoid anandamide or with the NPY1 receptor agonist Leu31,Pro34-NPY (Leu). Intra-BLA URB597 prevented the shock/reminders-induced PTSD- behaviors (extinction, startle) and depression-behaviors (despair, social impairments). These preventing effects of URB597 on PTSD- and depression-like behaviors were shown to be mostly mediated by cannabinoid CB1 and NPY1 receptors, as they were blocked when URB597 was co-administered with a low dose of a CB1 or NPY1 receptor antagonist. Similarly, intra-BLA Leu prevented development of all the behaviors. Interestingly, a CB1 antagonist prevented the effects of Leu on despair and social behavior, but not the effects on extinction and startle. Moreover, exposure to shock and reminders upregulated CB1 and NPY1 receptors in the BLA and infralimbic prefrontal cortex and this upregulation was restored to normal with intra-BLA URB597 or Leu. The findings suggest that the functional interaction between the eCB and NPY1 systems is complex and provide a rationale for exploring novel therapeutic strategies that target the cannabinoid and NPY systems for stress-related diseases.
Collapse
Affiliation(s)
- Neta Maymon
- Department of Psychology, University of Haifa, Haifa, 3498838, Israel
| | | | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College Valhalla, New York, 10595, USA
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
56
|
Zenz G, Farzi A, Fröhlich EE, Reichmann F, Holzer P. Intranasal Neuropeptide Y Blunts Lipopolysaccharide-Evoked Sickness Behavior but Not the Immune Response in Mice. Neurotherapeutics 2019; 16:1335-1349. [PMID: 31338703 PMCID: PMC6985076 DOI: 10.1007/s13311-019-00758-9] [Citation(s) in RCA: 6] [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] [Indexed: 01/23/2023] Open
Abstract
Neuropeptide Y (NPY) has been demonstrated to exert stress buffering effects and promote resilience. Non-invasive intranasal (IN) application of NPY to rodents is able to mitigate traumatic stress-induced behavioral changes as well as dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis. However, it is unknown whether IN NPY could prevent the behavioral, pro-inflammatory and neurochemical responses to peripheral immune activation by the Toll-like receptor 4 (TLR4) stimulant lipopolysaccharide (LPS). Therefore, we analyzed the effects of IN NPY (100 μg) on the behavioral sickness response (reduced locomotion and exploration) and the underlying molecular mechanisms, 3 h and 21 h after intraperitoneal injections of LPS (0.03 mg/kg) in male C57BL/6N mice. The acute behavioral sickness response was significantly dampened by pretreatment with IN NPY 3 h after LPS injection. This effect was accompanied by diminished weight loss and lowered plasma corticosterone (CORT) levels 21 h after LPS injection. In contrast, acute circulating cytokine levels and hypothalamic cytokine mRNA expression remained unaltered by IN NPY, which indicates that the peripheral and cerebral immune response to LPS was left undisturbed. Our findings are in agreement with the reported activity of NPY to dampen the response of the HPA axis to stress. We propose that IN NPY ablates sickness behavior at a site beyond the peripheral and cerebral cytokine response, an action that is associated with reduced activity of the HPA axis as determined by decreased plasma CORT.These results indicate that IN NPY administration may be relevant to the management of neuropsychiatric disorders arising from immune-induced neuroendocrine dysfunction.
Collapse
Affiliation(s)
- Geraldine Zenz
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria.
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
| | - Esther E Fröhlich
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Universitätsplatz 4, A-8010, Graz, Austria
- BioTechMed-Graz, Mozartgasse 12, A-8010, Graz, Austria
| |
Collapse
|
57
|
Seo JH, Park HS, Park SS, Kim CJ, Kim DH, Kim TW. Physical exercise ameliorates psychiatric disorders and cognitive dysfunctions by hippocampal mitochondrial function and neuroplasticity in post-traumatic stress disorder. Exp Neurol 2019; 322:113043. [PMID: 31446079 DOI: 10.1016/j.expneurol.2019.113043] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 07/24/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a stress-related condition that can be triggered by witnessing or experiencing a life-threatening event, such as a war, natural disaster, terrorist attack, major accident, or assault. PTSD is caused by dysfunction of the hippocampus and causes problems associated with brain functioning, such as anxiety, depression, and cognitive impairment. Exercise is known to have a positive effect on brain function, especially in the hippocampus. In this study, we investigated the effect of aerobic exercise on mitochondrial function and neuroplasticity in the hippocampus as well as behavioral changes in animal models of PTSD. Exposure to severe stress resulted in mitochondrial dysfunction in the hippocampus, including impaired Ca2+ homeostasis, an increase in reactive oxygen species such as H2O2, a decrease in the O2 respiration rate, and overexpression of membrane permeability transition pore-related proteins, including voltage-dependent anion channel, adenine nucleotide translocase, and cyclophilin-D. Exposure to extreme stress also decreased neuroplasticity by increasing apoptosis and decreasing the brain-derived neurotrophic factor level and neurogenesis, resulting in increased anxiety, depression, and cognitive impairment. The impairments in mitochondrial function and neuroplasticity in the hippocampus, as well as anxiety, depression, and cognitive impairment, were all improved by exercise. Exercise-induced improvement of the brain-derived neurotrophic factor level in particular might alter mitochondrial function, neuroplasticity, and the rate of apoptosis in the hippocampus. Therefore, exercise might be an important non-pharmacological intervention for the prevention and treatment of the pathobiology of PTSD.
Collapse
Affiliation(s)
- Jin-Hee Seo
- Department of Adapted physical education, Baekseok University, Cheonan, Republic of Korea
| | - Hye-Sang Park
- Department of Kinesiology, College of public health and Cardiovascular Research Center, Lewis Katz school of Medicine, Temple University, Philadelphia, PA, USA
| | - Sang-Seo Park
- Department of physiology, College of medicine, Kyung Hee University, Seoul, Republic of Korea; Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Chang-Ju Kim
- Department of physiology, College of medicine, Kyung Hee University, Seoul, Republic of Korea; Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Dong-Hyun Kim
- College of Sports science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tae-Woon Kim
- Department of physiology, College of medicine, Kyung Hee University, Seoul, Republic of Korea; Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea; Exercise Rehabilitation Research Institute, Department of Exercise & Health Science, Sangmyung University, Seoul, Republic of Korea.
| |
Collapse
|
58
|
Sun R, Liu Y, Hou B, Lei Y, Bo J, Zhang W, Sun Y, Zhang Y, Zhang Z, Liu Z, Huo W, Mao Y, Ma Z, Gu X. Perioperative activation of spinal α7 nAChR promotes recovery from preoperative stress-induced prolongation of postsurgical pain. Brain Behav Immun 2019; 79:294-308. [PMID: 30797046 DOI: 10.1016/j.bbi.2019.02.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 01/20/2019] [Accepted: 02/20/2019] [Indexed: 12/01/2022] Open
Abstract
Preoperative stress could delay the recovery of postoperative pain and has been reported to be a risk factor for chronic postsurgical pain. As stress could facilitate the proinflammatory activation of microglia, we hypothesized that these cells may play a vital role in the development of preoperative stress-induced pain chronification after surgery. Our experiments were conducted in a rat model that consists of a single prolonged stress (SPS) procedure and plantar incision. A previous SPS exposure induced anxiety-like behaviors, prolonged incision-induced mechanical allodynia, and potentiated the activation of spinal microglia. Based on the results from ex vivo experiments, spinal microglia isolated from SPS-exposed rats secreted more proinflammatory cytokines upon challenge with LPS. Our results also demonstrated that microglia played a more important role than astrocytes in the initiation of SPS-induced prolongation of postsurgical pain. We further explored the therapeutic potential of agonism of α7 nAChR, an emerging anti-inflammatory target, for SPS-induced prolongation of postsurgical pain. Multiple intrathecal (i.t.) injections of PHA-543613 (an α7 nAChR agonist) or PNU-120596 (a type II positive allosteric modulator) during the perioperative period shortened the duration of postsurgical pain after SPS and suppressed SPS-potentiated microglia activation, but their effects were abolished by pretreatment with methyllycaconitine (an α7 nAChR antagonist; i.t.). Based on the results from ex vivo experiments, the anti-inflammatory effects of PHA-543613 and PNU-120596 may have been achieved by the direct modulation of microglia. In conclusion, stress-induced priming of spinal microglia played a key role in the initiation of preoperative stress-induced prolongation of postsurgical pain, and PHA-543613 and PNU-120596 may be potential candidates for preventing pain chronification after surgery.
Collapse
Affiliation(s)
- Rao Sun
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Liu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Bailing Hou
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yishan Lei
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Jinhua Bo
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Wei Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yu'E Sun
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Ying Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Zuoxia Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Zhe Liu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Wenwen Huo
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yanting Mao
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Zhengliang Ma
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.
| |
Collapse
|
59
|
Cheon M, Park H, Rhim H, Chung C. Actions of Neuropeptide Y on Synaptic Transmission in the Lateral Habenula. Neuroscience 2019; 410:183-190. [PMID: 31082535 DOI: 10.1016/j.neuroscience.2019.04.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/27/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022]
Abstract
Neuropeptide Y is a peptide neuromodulator with protective roles including anxiolytic and antidepressant-like effects in animal models of depression and post-traumatic stress disorder. The lateral habenula (LHb) is a brain region that encodes aversive information and is closely related with mood disorders. Although LHb neurons express NPY receptors, the physiological roles of NPY in this region remain uninvestigated. In this study, we examined the actions of NPY on synaptic transmission in the LHb using whole cell patch clamp recording. We observed that NPY inhibited excitatory neurotransmission in a subset of LHb neurons whereas potentiating in a small population of neurons. Inhibitory transmission remained unchanged by NPY application in a subset of neurons but was reduced in the majority of LHb neurons recorded. The overall outcome of NPY application was a decrease in the spontaneous firing rate of the LHb, leading to hypoactivation of the LHb. Our observations indicate that although NPY has divergent effects on excitatory and inhibitory transmission, NPY receptor activation decreases LHb activity, suggesting that the LHb may partly mediate the protective roles of NPY in the central nervous system.
Collapse
Affiliation(s)
- Myunghyun Cheon
- Department of Biological Sciences, Konkuk University, Seoul 05029, South Korea
| | - Hoyong Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, South Korea
| | - Hyewon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 139-791, South Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul 05029, South Korea.
| |
Collapse
|
60
|
Nwokafor C, Serova LI, Sabban EL. Preclinical findings on the potential of intranasal neuropeptide Y for treating hyperarousal features of PTSD. Ann N Y Acad Sci 2019; 1455:149-159. [PMID: 31250475 DOI: 10.1111/nyas.14172] [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: 02/18/2019] [Revised: 05/20/2019] [Accepted: 05/29/2019] [Indexed: 01/10/2023]
Abstract
Acoustic startle response (ASR) assesses hyperarousal, a core symptom of posttraumatic stress disorder (PTSD). Intranasal neuropeptide Y (NPY) administration was shown to prevent hyperarousal in single prolonged stress (SPS) rodent PTSD model. However, it is unclear how ASR itself alters responses to stress. Rats (A-S-A) were exposed to acoustic startle (AS) 1 day before SPS (ASR1) and 2 weeks afterward (ASR2). Other groups were exposed in parallel to either AS (A-A) or SPS or neither. SPS enhanced ASR2. In relevant brain areas, mRNA levels were determined by qRT-PCR. In mediobasal hypothalamus, AS or SPS each increased CRH mRNA levels without an additive effect. Exposure to AS appeared to dampen some responses to SPS. The SPS-triggered reduction of GR and FKBP5 gene expression was not observed in A-S-A group. In locus coeruleus, SPS increased CRHR1 and reduced Y2R mRNAs, but not in A-S-A group. In both regions, AS altered NPY receptor gene expression, which may mediate dampening responses to SPS. In second experiment, intranasal NPY administered 2 weeks after SPS reversed hyperarousal symptoms for at least 7 days. This study reveals important effects of AS on the NPY system and demonstrates that intranasal NPY elicits long-lasting reversal of traumatic stress-triggered hyperarousal.
Collapse
Affiliation(s)
- Chiso Nwokafor
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| |
Collapse
|
61
|
Miles OW, Maren S. Role of the Bed Nucleus of the Stria Terminalis in PTSD: Insights From Preclinical Models. Front Behav Neurosci 2019; 13:68. [PMID: 31024271 PMCID: PMC6461014 DOI: 10.3389/fnbeh.2019.00068] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/18/2019] [Indexed: 12/18/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) afflicts approximately 8% of the United States population and represents a significant public health burden, but the underlying neural mechanisms of this and other anxiety- and stressor-related disorders are largely unknown. Within the last few decades, several preclinical models of PSTD have been developed to help elucidate the mechanisms underlying dysregulated fear states. One brain area that has emerged as a critical mediator of stress-related behavioral processing in both clinical and laboratory settings is the bed nucleus of the stria terminalis (BNST). The BNST is interconnected with essential emotional processing regions, including prefrontal cortex, hippocampus and amygdala. It is activated by stressor exposure and undergoes neurochemical and morphological alterations as a result of stressor exposure. Stress-related neuro-peptides including corticotropin-releasing factor (CRF) and pituitary adenylate cyclase activating peptide (PACAP) are also abundant in the BNST, further implicating an involvement of BNST in stress responses. Behaviorally, the BNST is critical for acquisition and expression of fear and is well positioned to regulate fear relapse after periods of extinction. Here, we consider the role of the BNST in stress and memory processes in the context of preclinical models of PTSD.
Collapse
Affiliation(s)
- Olivia W. Miles
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | | |
Collapse
|
62
|
Serova LI, Nwokafor C, Van Bockstaele EJ, Reyes BAS, Lin X, Sabban EL. Single prolonged stress PTSD model triggers progressive severity of anxiety, altered gene expression in locus coeruleus and hypothalamus and effected sensitivity to NPY. Eur Neuropsychopharmacol 2019; 29:482-492. [PMID: 30878321 DOI: 10.1016/j.euroneuro.2019.02.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/17/2022]
Abstract
PTSD is heterogeneous disorder that can be long lasting and often has delayed onset following exposure to a traumatic event. Therefore, it is important to take a staging approach to evaluate progression of biological mechanisms of the disease. Here, we begin to evaluate the temporal trajectory of changes following exposure to traumatic stressors in the SPS rat PTSD model. The percent of animals displaying severe anxiety on EPM increased from 17.5% at one week to 57.1% two weeks after SPS stressors, indicating delayed onset or progressive worsening of the symptoms. The LC displayed prolonged activation, and dysbalance of the CRH/NPY systems, with enhanced CRHR1 gene expression, coupled with reduced mRNAs for NPY and Y2R. In the mediobasal hypothalamus, increased CRH mRNA levels were sustained, but there was a flip in alterations of HPA regulatory molecules, GR and FKBP5 and Y5 receptor at two weeks compared to one week. Two weeks after SPS, intranasal NPY at 300 µg/rat, but not 150 µg which was effective after one week, reversed SPS triggered elevated anxiety. It also reversed SPS elicited depressive/despair symptoms and hyperarousal. Overall, the results reveal time-dependent progression in development of anxiety symptoms and molecular impairments in gene expression for CRH and NPY systems in LC and mediobasal hypothalamus by SPS. With longer time afterwards only a higher dose of NPY was effective in reversing behavioral impairments triggered by SPS, indicating that therapeutic approaches should be adjusted according to the degree of biological progression of the disorder.
Collapse
Affiliation(s)
- Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College Valhalla, Basic Sciences Building, New York, NY 10595, USA
| | - Chiso Nwokafor
- Department of Biochemistry and Molecular Biology, New York Medical College Valhalla, Basic Sciences Building, New York, NY 10595, USA
| | | | - Beverly A S Reyes
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, PA 19012, USA
| | - Xiaoping Lin
- Department of Biochemistry and Molecular Biology, New York Medical College Valhalla, Basic Sciences Building, New York, NY 10595, USA
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College Valhalla, Basic Sciences Building, New York, NY 10595, USA.
| |
Collapse
|
63
|
Sabban EL, Serova LI. Potential of Intranasal Neuropeptide Y (NPY) and/or Melanocortin 4 Receptor (MC4R) Antagonists for Preventing or Treating PTSD. Mil Med 2019; 183:408-412. [PMID: 29635611 DOI: 10.1093/milmed/usx228] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/18/2018] [Indexed: 02/02/2023] Open
Abstract
There is a great need for effective treatment options for post-traumatic stress disorder (PTSD). Neuropeptide Y (NPY) is associated with resilience to traumatic stress. MC4R antagonists, such as HS014, also reduce response to stress. Both regulate stress-responsive systems - the hypothalamic-pituitary-axis (HPA) and the noradrenergic nervous system and their associated behaviors. Therefore, we examined if their intranasal delivery to brain could attenuate development of PTSD-related symptoms in single prolonged stress (SPS) rodent PTSD model. Three regimens were used: (1) prophylactic treatment 30 min before SPS stressors, (2) early intervention right after SPS stressors, (3) therapeutic treatment when PTSD behaviors are manifested 1 wk or more after the traumatic stress. NPY delivered by regimen 1 or 2 prevented SPS-triggered elevation in anxiety, depressive-like behavior, and hyperarousal and reduced dysregulation of HPA axis. Hypothalamic CRH mRNA and GR in ventral hippocampus were significantly induced in vehicle- but not NPY-treated group. NPY also prevented hypersensitivity of LC/NE system to novel mild stressor and induction of CRH in amygdala. Some of these impairments were also reduced with HS014, alone or together with NPY. When given after symptoms were manifested (regiment 3), NPY attenuated anxiety and depressive behaviors. This demonstrates strong preclinical proof of concept for intranasal NPY, and perhaps MC4R antagonists, for non-invasive early pharmacological interventions for PTSD and comorbid disorders and possibly also as therapeutic strategy.
Collapse
Affiliation(s)
- Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595
| |
Collapse
|
64
|
Tangthavewattana S, Leelawatwattana L, Prapunpoj P. The hydrophobic C-terminal sequence of transthyretin affects its catalytic kinetics towards amidated neuropeptide Y. FEBS Open Bio 2019; 9:594-604. [PMID: 30984535 PMCID: PMC6443995 DOI: 10.1002/2211-5463.12604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/20/2018] [Indexed: 12/03/2022] Open
Abstract
Transthyretin (TTR) is a transporter for thyroid hormone and retinol binding protein that has recently been reported to have proteolytic activity against certain substrates, including amidated neuropeptide Y (NPY). However, the proteolytic activity of TTR towards NPY is not fully understood. Here, we used fluorescence-based assays to determine the catalytic kinetics of human TTR towards human amidated NPY. The Michaelis constant (KM) and catalytic efficiency (kcat/KM) of TTR proteolysis were 15.88 ± 0.44 μm and 687 081 ± 35 692 m -1·s-1, respectively. In addition, we demonstrated an effect of the C-terminal sequence of TTR. When the C-terminal sequence of TTR was made more hydrophobic, the KM and kcat/KM changed to 12.87 ± 0.22 μm and 983 755 ± 18 704 m -1·s-1, respectively. Our results may be useful for the development of TTR as a therapeutic agent with low risk of the undesirable symptoms that develop from amidated NPY, and for further improvement of the kcat/KM of TTR.
Collapse
Affiliation(s)
| | - Ladda Leelawatwattana
- Department of BiochemistryFaculty of SciencePrince of Songkla UniversityHat YaiThailand
| | - Porntip Prapunpoj
- Department of BiochemistryFaculty of SciencePrince of Songkla UniversityHat YaiThailand
| |
Collapse
|
65
|
Abstract
This review examines the putative link between glucocorticoid and hippocampal abnormalities in posttraumatic stress disorder (PTSD). Increased glucocorticoid receptor (GR) sensitivity in PTSD may permit enhanced negative feedback inhibition of cortisol at the pituitary, hypothalamus, or other brain regions comprising the hypothalamic-pituitary-adrenal (HPA) axis and would be expected to affect other physiological systems that are regulated by glucocorticoids. Molecular and transcriptional studies of cortisol are consistent with the hypothesis that cortisol actions may be amplified in PTSD as a result of enhanced GR sensitivity in monocytes and some brain regions, although cortisol levels themselves are unchanged and oftentimes lower than normal. Concurrently, magnetic resonance imaging studies have demonstrated that individuals with PTSD have smaller hippocampal volume than individuals without PTSD. Initial hypotheses regarding the mechanism underlying hippocampal alterations in PTSD focused on elevated glucocorticoid levels in combination with extreme stress as the primary cause, but this explanation has not been well supported in human studies. Lack of data from neuroimaging studies preclude a firm link between PTSD onset and hippocampal volume changes. Rather, the available evidence is consistent with the possibility that smaller hippocampal volume (like reduced cortisol levels and enhanced GR sensitivity) may be a vulnerability factor for developing the disorder; limitations of hippocampal-based models of PTSD are described. We further review neuroimaging studies examining hippocampal structure and function following manipulation of glucocorticoid levels and also examining changes in the hippocampus in relationship to other brain regions. Evidence that the GR may be an important therapeutic target for the treatment of PTSD, especially for functions subserved by the hippocampus, is discussed. Implications of the current review for future research are described, with an emphasis on the need to integrate findings of glucocorticoid abnormalities with functional-imaging paradigms to formulate a comprehensive model of HPA-axis functioning in PTSD.
Collapse
|
66
|
Abstract
Resilience is defined as the dynamic ability to adapt successfully in the face of adversity, trauma, or significant threat. Some of the key early studies of resilience were observational studies in children. They were followed by research in adults, studies testing interventions to promote resilience in different populations, and a recent upsurge of studies on the underlying genomic and neurobiological mechanisms. Neural and molecular studies in preclinical models of resilience are also increasingly identifying active stress adaptations in resilient animals. Knowledge gained from animal and human studies of resilience can be harnessed to develop new preventive interventions to enhance resilience in at-risk populations. Further, treatment interventions focused on enhancing potentially modifiable protective factors that are consistently linked to psychological resilience can enrich currently available treatment interventions for individuals with posttraumatic stress disorder (PTSD). Translating our expanding knowledge of the neurobiology of resilience additionally promises to yield novel therapeutic strategies for treating this disabling condition. This review summarizes the vast field of resilience research spanning genomic, psychosocial, and neurobiological levels, and discusses how findings have led and can lead to new preventive and treatment interventions for PTSD.
Collapse
|
67
|
Joksimovic J, Selakovic D, Jovicic N, Mitrovic S, Mihailovic V, Katanic J, Milovanovic D, Rosic G. Exercise Attenuates Anabolic Steroids-Induced Anxiety via Hippocampal NPY and MC4 Receptor in Rats. Front Neurosci 2019; 13:172. [PMID: 30863280 PMCID: PMC6399386 DOI: 10.3389/fnins.2019.00172] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/13/2019] [Indexed: 12/16/2022] Open
Abstract
The aim of our study was to evaluate the effects of chronic administration of nandrolone-decanoate (ND) or testosterone-enanthate (TE) in supraphysiological doses and a prolonged swimming protocol, alone and in combination with ND or TE, on anxiety-like behavior in rats. We investigated the immunohistochemical alterations of the hippocampal neuropeptide Y (NPY) and melanocortin 4 receptor (MC4R) neurons, as a possible underlying mechanism in a modulation of anxiety-like behavior in rats. Both applied anabolic androgenic steroids (AASs) induced anxiogenic effect accompanied with decreased serum and hippocampal NPY. The exercise-induced anxiolytic effect was associated with increased hippocampal NPY expression. ND and TE increased the number of MC4R, while the swimming protocol was followed by the reduction of MC4R in the CA1 region of the hippocampus. However, NPY/MC4R ratio in hippocampus was lowered by AASs and elevated by exercise in all hippocampal regions. An augmentation of this ratio strongly and positively correlated to increased time in open arms of elevated plus maze, in the context that indicates anxiolytic effect. Our findings support the conclusion that alterations in both hippocampal NPY and MC4R expression are involved in anxiety level changes in rats, while their quantitative relationship (NPY/MC4R ratio) is even more valuable in the estimation of anxiety regulation than individual alterations for both NPY and MC4R expression in the hippocampus.
Collapse
Affiliation(s)
- Jovana Joksimovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Slobodanka Mitrovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladimir Mihailovic
- Department of Chemistry, Faculty of Science, University of Kragujevac, Kragujevac, Serbia
| | - Jelena Katanic
- Department of Chemistry, Faculty of Science, University of Kragujevac, Kragujevac, Serbia
| | - Dragan Milovanovic
- Department of Pharmacology and Toxicology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gvozden Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
68
|
Nahvi RJ, Nwokafor C, Serova LI, Sabban EL. Single Prolonged Stress as a Prospective Model for Posttraumatic Stress Disorder in Females. Front Behav Neurosci 2019; 13:17. [PMID: 30804766 PMCID: PMC6378310 DOI: 10.3389/fnbeh.2019.00017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/21/2019] [Indexed: 12/27/2022] Open
Abstract
Sex plays an important role in susceptibility to stress triggered disorders. Posttraumatic Stress disorder (PTSD), a debilitating psychiatric disorder developed after exposure to a traumatic event, is two times more prevalent in women than men. However, the vast majority of animal models of PTSD, including single prolonged stress (SPS), were performed mostly with males. Here, we evaluated SPS as an appropriate PTSD model for females in terms of anxiety, depressive symptoms and changes in gene expression in the noradrenergic system in the brain. In addition, we examined intranasal neuropeptide Y (NPY) as a possible treatment in females. Female rats were subjected to SPS and given either intranasal NPY or vehicle in two separate experiments. In the first experiment, stressed females were compared to unstressed controls on forced swim test (FST) and for levels of expression of several genes in the locus coeruleus (LC) 12 days after SPS exposure. Using a separate cohort of animals, experiment two examined stressed females and unstressed controls on the elevated plus maze (EPM) and LC gene expression 7 days after SPS stressors. SPS led to increased anxiety-like behavior on EPM and depressive-like behavior on FST. Following FST, the rats displayed elevated tyrosine hydroxylase (TH), CRHR1 and Y1R mRNA levels in the LC, consistent with increased activation of the noradrenergic system. The expression level of these mRNAs was unchanged following EPM, except Y1R. Intranasal NPY at the doses shown to be effective in males, did not prevent development of depressive or anxiety-like behavior or molecular changes in the LC. The results indicate that while SPS could be an appropriate PTSD model for females, sex differences, such as response to NPY, are important to consider.
Collapse
Affiliation(s)
- Roxanna J Nahvi
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Chiso Nwokafor
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
69
|
Golub Y, Schildbach EM, Touma C, Kratz O, Moll GH, von Hörsten S, Canneva F. Role of hypothalamus-pituitary-adrenal axis modulation in the stress-resilient phenotype of DPP4-deficient rats. Behav Brain Res 2019; 356:243-249. [DOI: 10.1016/j.bbr.2018.08.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 01/30/2023]
|
70
|
Manukhina EB, Tseilikman VE, Tseilikman OB, Komelkova MV, Kondashevskaya MV, Goryacheva AV, Lapshin MS, Platkovskii PO, Alliluev AV, Downey HF. Intermittent hypoxia improves behavioral and adrenal gland dysfunction induced by posttraumatic stress disorder in rats. J Appl Physiol (1985) 2018; 125:931-937. [DOI: 10.1152/japplphysiol.01123.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nonpharmacological treatments of stress-induced disorders are promising, since they enhance endogenous stress defense systems, are free of side effects, and have few contraindications. The present study tested the hypothesis that intermittent hypoxia conditioning (IHC) ameliorates behavioral, biochemical, and morphological signs of experimental posttraumatic stress disorder (PTSD) induced in rats with a model of predator stress (10-day exposure to cat urine scent, 15 min daily followed by 14 days of stress-free rest). After the last day of stress exposure, rats were conditioned in an altitude chamber for 14 days at a 1,000-m simulated altitude for 30 min on day 1 with altitude and duration progressively increasing to 4,000 m for 4 h on day 5. PTSD was associated with decreased time spent in open arms and increased time spent in closed arms of the elevated X-maze, increased anxiety index, and increased rate of freezing responses. Functional and structural signs of adrenal cortex degeneration were also observed, including decreased plasma concentration of corticosterone, decreased weight of adrenal glands, reduced thickness of the fasciculate zone, and hydropic degeneration of adrenal gland cells. The thickness of the adrenal fasciculate zone negatively correlated with the anxiety index. IHC alleviated both behavioral signs of PTSD and morphological evidence of adrenal cortex dystrophy. Also, IHC alone exerted an antistress effect, which was evident from the increased time spent in open arms of the elevated X-maze and a lower number of rats displaying freezing responses. Therefore, IHC of rats with experimental PTSD reduced behavioral signs of the condition and damage to the adrenal glands. NEW & NOTEWORTHY Intermittent hypoxia conditioning (IHC) has been shown to be cardio-, vaso-, and neuroprotective. For the first time, in a model of posttraumatic stress disorder (PTSD), this study showed that IHC alleviated both PTSD-induced behavioral disorders and functional and morphological damage to the adrenal glands. Also, IHC alone exerted an antistress effect. These results suggest that IHC may be a promising complementary treatment for PTSD-associated disorders.
Collapse
Affiliation(s)
- Eugenia B. Manukhina
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
- Laboratory for Regulatory Mechanisms of Stress and Adaptation, Institute of General Pathology and Pathophysiology, Moscow, Russia
- Department of Anatomy and Physiology, University of North Texas Health Science Center, Fort Worth, Texas
| | | | - Olga B. Tseilikman
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Maria V. Komelkova
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Marina V. Kondashevskaya
- Laboratory for Immunomorphology of Inflammation, FSBSI Research Institute of Human Morphology, Moscow, Russia
| | - Anna V. Goryacheva
- Laboratory for Regulatory Mechanisms of Stress and Adaptation, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Maxim S. Lapshin
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | | | | | - H. Fred Downey
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
- Department of Anatomy and Physiology, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
71
|
Liu H, Zhang C, Ji Y, Yang L. Biological and Psychological Perspectives of Resilience: Is It Possible to Improve Stress Resistance? Front Hum Neurosci 2018; 12:326. [PMID: 30186127 PMCID: PMC6110926 DOI: 10.3389/fnhum.2018.00326] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 07/25/2018] [Indexed: 12/23/2022] Open
Abstract
The term “resilience” refers to the ability to adapt successfully to stress, trauma and adversity, enabling individuals to avoid stress-induced mental disorders such as depression, posttraumatic stress disorder (PTSD) and anxiety. Here, we review evidence from both animal models and humans that is increasingly revealing the neurophysiological and neuropsychological mechanisms that underlie stress susceptibility, as well as active mechanisms underlying the resilience phenotype. Ultimately, this growing understanding of the neurobiological mechanisms of resilience should result in the development of novel interventions that specifically target neural circuitry and brain areas that enhance resilience and lead to more effective treatments for stress-induced disorders. Stress resilience can be improved, but the outcomes and effects depend on the type of intervention and the species treated.
Collapse
Affiliation(s)
- Haoran Liu
- School of Psychology, Center for Studies of Psychological Application, South China Normal University, Guangzhou, China
| | - Chenfeng Zhang
- School of Psychology, Center for Studies of Psychological Application, South China Normal University, Guangzhou, China
| | - Yannan Ji
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Li Yang
- School of Psychology, Center for Studies of Psychological Application, South China Normal University, Guangzhou, China.,Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| |
Collapse
|
72
|
Oh JY, Kim YK, Kim SN, Lee B, Jang JH, Kwon S, Park HJ. Acupuncture modulates stress response by the mTOR signaling pathway in a rat post-traumatic stress disorder model. Sci Rep 2018; 8:11864. [PMID: 30089868 PMCID: PMC6082850 DOI: 10.1038/s41598-018-30337-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 07/25/2018] [Indexed: 01/02/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disease that can form following exposure to a traumatic event. Acupuncture has been proposed as a beneficial treatment for PTSD, but the underlying mechanisms remain unclear. The present study investigated whether acupuncture improves depression- and anxiety-like behaviors induced using a single prolonged stress (SPS) as a PTSD rat model. In addition, we investigated whether the effects were mediated by increased mTOR activity and its downstream signaling components, which contribute to protein synthesis required for synaptic plasticity in the hippocampus. We found that acupuncture at HT8 significantly alleviated both depression- and anxiety-like behaviors induced by SPS in rats, as assessed by the forced swimming, elevated plus maze, and open field tests; this alleviation was blocked by rapamycin. The effects of acupuncture were equivalent to those exerted by fluoxetine. Acupuncture regulated protein translation in the mTOR signaling pathway and enhanced the activation of synaptic proteins, PSD95, Syn1, and GluR1 in the hippocampus. These results suggest that acupuncture exerts antidepressant and anxiolytic effects on PTSD-related symptoms by increasing protein synthesis required for synaptic plasticity via the mTOR pathway in the hippocampus. Acupuncture may be a promising treatment for patients with PTSD and play a role as an alternative PTSD treatment.
Collapse
Affiliation(s)
- Ju-Young Oh
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea
| | - Yu-Kang Kim
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea
| | - Seung-Nam Kim
- College of Korean Medicine, Dongguk University, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Jae-Hwan Jang
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea
| | - Sunoh Kwon
- Korean Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea. .,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea. .,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
73
|
Miragaia AS, de Oliveira Wertheimer GS, Consoli AC, Cabbia R, Longo BM, Girardi CEN, Suchecki D. Maternal Deprivation Increases Anxiety- and Depressive-Like Behaviors in an Age-Dependent Fashion and Reduces Neuropeptide Y Expression in the Amygdala and Hippocampus of Male and Female Young Adult Rats. Front Behav Neurosci 2018; 12:159. [PMID: 30131681 PMCID: PMC6090069 DOI: 10.3389/fnbeh.2018.00159] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/09/2018] [Indexed: 01/28/2023] Open
Abstract
Maternal deprivation for 24 h produces an immediate increase in basal and stress-induced corticosterone (CORT) secretion. Given the impact of elevated CORT levels on brain development, the goal of the present study was to characterize the effects of maternal deprivation at postnatal days 3 (DEP3) or 11 (DEP11) on emotional behavior and neuropeptide Y immunoreactivity (NPY-ir) in the basolateral amygdala (BLA) and dorsal hippocampus (dHPC) of male and female rats. Litters were distributed in control non-deprived (CTL), DEP3, or DEP11 groups. In Experiment 1, within each litter, one male and one female were submitted to one of the following tests: novelty suppressed feeding (NSF), sucrose negative contrast test (SNCT), and forced swimming test (FST), between postnatal days 52 and 60. In Experiment 2, two males and two females per litter were exposed to the elevated plus maze and 1 h later, perfused for investigation of NPY-ir, on PND 52. The results showed that DEP3 rats displayed greater anxiety-like behavior in the NSF and EPM, compared to CTL and DEP11 counterparts. In the SNCT, DEP3 and DEP11 males showed less suppression of the lower sucrose concentration intake, whereas all females suppressed less than males. Both manipulated groups displayed more immobility in the FST, although this effect was greater in DEP3 than in DEP11 rats. NPY-ir was reduced in DEP3 and DEP11 males and females in the BLA, whereas in the dHPC, DEP3 males showed less NPY-ir than DEP11, which, in turn, presented less NPY-ir than CTL rats. Females showed less NPY-ir than males in both structures. Because the deprivation effects were more intense in DEP3 than in DEP11, in Experiment 3, the frequency of nursing posture, licking-grooming, and interaction with pups was assessed upon litter reunion with mothers. Mothers of DEP11 litters engaged more in anogenital licking than mothers of DEP3 litters. The present results indicate that maternal deprivation changed affective behavior with greater impact in the earlier age and reduced the expression of NPY in emotion-related brain areas. The age-dependent differential effects of deprivation on maternal behavior could, at least in part, explain the outcomes in young adult rats.
Collapse
Affiliation(s)
- Alexandra S Miragaia
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Amanda C Consoli
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rafael Cabbia
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Beatriz M Longo
- Departamento de Fisiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carlos E N Girardi
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deborah Suchecki
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
74
|
Fitzpatrick CJ, Jagannathan L, Lowenstein ED, Robinson TE, Becker JB, Morrow JD. Single prolonged stress decreases sign-tracking and cue-induced reinstatement of cocaine-seeking. Behav Brain Res 2018; 359:799-806. [PMID: 30077578 DOI: 10.1016/j.bbr.2018.07.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/27/2018] [Accepted: 07/27/2018] [Indexed: 11/19/2022]
Abstract
Exposure to prolonged, uncontrollable stress reduces reward-seeking behavior, resulting in anhedonia in neuropsychiatric disorders, such as posttraumatic stress disorder. However, it is unclear to what degree stressed subjects lose interest in rewards themselves or in reward-related cues that instigate reward-seeking behavior. In the present study, we investigated the effects of single prolonged stress (SPS) on cue-directed behavior in two different procedures: Pavlovian conditioned approach (PCA) and cue-induced reinstatement of cocaine-seeking. In Experiment 1, rats were exposed to SPS and tested for the acquisition of sign-tracking (cue-directed) and goal-tracking (reward-directed) behaviors during a PCA procedure. In Experiment 2, rats were exposed to SPS and tested for the expression of sign- and goal-tracking as well as cue-induced reinstatement of cocaine-seeking. Because dopaminergic activity in the nucleus accumbens is known to play a central role in many cue-directed behaviors, including both sign-tracking and cue-induced reinstatement, Experiment 3 used in vivo microdialysis to measure the effect of SPS on baseline and evoked dopamine levels in the nucleus accumbens. SPS decreased sign-tracking and increased goal-tracking during the acquisition of PCA behavior without affecting reward consumption. In addition, SPS decreased cue-induced reinstatement without affecting cocaine self-administration. Finally, SPS decreased evoked but not baseline levels of dopamine in the nucleus accumbens. These results suggest that SPS decreases the motivational, but not consummatory, aspects of reward-seeking behavior, which may result from long-term, SPS-induced reductions in dopamine release in the nucleus accumbens.
Collapse
Affiliation(s)
| | | | | | - Terry E Robinson
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jill B Becker
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan D Morrow
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
75
|
Theisen CC, Reyes BA, Sabban E, Van Bockstaele EJ. Ultrastructural Characterization of Corticotropin-Releasing Factor and Neuropeptide Y in the Rat Locus Coeruleus: Anatomical Evidence for Putative Interactions. Neuroscience 2018; 384:21-40. [DOI: 10.1016/j.neuroscience.2018.04.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022]
|
76
|
Prefrontal cortex-dependent innate behaviors are altered by selective knockdown of Gad1 in neuropeptide Y interneurons. PLoS One 2018; 13:e0200809. [PMID: 30024942 PMCID: PMC6053188 DOI: 10.1371/journal.pone.0200809] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 06/09/2018] [Indexed: 12/23/2022] Open
Abstract
GABAergic dysfunction has been implicated in a variety of neurological and psychiatric disorders, including anxiety disorders. Anxiety disorders are the most common type of psychiatric disorder during adolescence. There is a deficiency of GABAergic transmission in anxiety, and enhancement of GABA transmission through pharmacological means reduces anxiety behaviors. GAD67—the enzyme responsible for GABA production–has been linked to anxiety disorders. One class of GABAergic interneurons, Neuropeptide Y (NPY) expressing cells, is abundantly found in brain regions associated with anxiety and fear learning, including prefrontal cortex, hippocampus and amygdala. Additionally, NPY itself has been shown to have anxiolytic effects, and loss of NPY+ interneurons enhances anxiety behaviors. A previous study showed that knockdown of Gad1 from NPY+ cells led to reduced anxiety behaviors in adult mice. However, the role of GABA release from NPY+ interneurons in adolescent anxiety is unclear. Here we used a transgenic mouse that reduces GAD67 in NPY+ cells (NPYGAD1-TG) through Gad1 knockdown and tested for effects on behavior in adolescent mice. Adolescent NPYGAD1-TG mice showed enhanced anxiety-like behavior and sex-dependent changes in locomotor activity. We also found enhancement in two other innate behavioral tasks, nesting construction and social dominance. In contrast, fear learning was unchanged. Because we saw changes in behavioral tasks dependent upon prefrontal cortex and hippocampus, we investigated the extent of GAD67 knockdown in these regions. Immunohistochemistry revealed a 40% decrease in GAD67 in NPY+ cells in prefrontal cortex, indicating a significant but incomplete knockdown of GAD67. In contrast, there was no decrease in GAD67 in NPY+ cells in hippocampus. Consistent with this, there was no change in inhibitory synaptic transmission in hippocampus. Our results show the behavioral impact of cell-specific interneuron dysfunction and suggest that GABA release by NPY+ cells is important for regulating innate prefrontal cortex-dependent behavior in adolescents.
Collapse
|
77
|
Neurotransmitter, Peptide, and Steroid Hormone Abnormalities in PTSD: Biological Endophenotypes Relevant to Treatment. Curr Psychiatry Rep 2018; 20:52. [PMID: 30019147 DOI: 10.1007/s11920-018-0908-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW This review summarizes neurotransmitter, peptide, and other neurohormone abnormalities associated with posttraumatic stress disorder (PTSD) and relevant to development of precision medicine therapeutics for PTSD. RECENT FINDINGS As the number of molecular abnormalities associated with PTSD across a variety of subpopulations continues to grow, it becomes clear that no single abnormality characterizes all individuals with PTSD. Instead, individually variable points of molecular dysfunction occur within several different stress-responsive systems that interact to produce the clinical PTSD phenotype. Future work should focus on critical interactions among the systems that influence PTSD risk, severity, chronicity, comorbidity, and response to treatment. Effort also should be directed toward development of clinical procedures by which points of molecular dysfunction within these systems can be identified in individual patients. Some molecular abnormalities are more common than others and may serve as subpopulation biological endophenotypes for targeting of currently available and novel treatments.
Collapse
|
78
|
Deslauriers J, Toth M, Der-Avakian A, Risbrough VB. Current Status of Animal Models of Posttraumatic Stress Disorder: Behavioral and Biological Phenotypes, and Future Challenges in Improving Translation. Biol Psychiatry 2018; 83:895-907. [PMID: 29338843 PMCID: PMC6085893 DOI: 10.1016/j.biopsych.2017.11.019] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/05/2017] [Accepted: 11/03/2017] [Indexed: 12/23/2022]
Abstract
Increasing predictability of animal models of posttraumatic stress disorder (PTSD) has required active collaboration between clinical and preclinical scientists. Modeling PTSD is challenging, as it is a heterogeneous disorder with ≥20 symptoms. Clinical research increasingly utilizes objective biological measures (e.g., imaging, peripheral biomarkers) or nonverbal behaviors and/or physiological responses to complement verbally reported symptoms. This shift toward more-objectively measurable phenotypes enables refinement of current animal models of PTSD, and it supports the incorporation of homologous measures across species. We reviewed >600 articles to examine the ability of current rodent models to probe biological phenotypes of PTSD (e.g., sleep disturbances, hippocampal and fear-circuit dysfunction, inflammation, glucocorticoid receptor hypersensitivity) in addition to behavioral phenotypes. Most models reliably produced enduring generalized anxiety-like or depression-like behaviors, as well as hyperactive fear circuits, glucocorticoid receptor hypersensitivity, and response to long-term selective serotonin reuptake inhibitors. Although a few paradigms probed fear conditioning/extinction or utilized peripheral immune, sleep, and noninvasive imaging measures, we argue that these should be incorporated more to enhance translation. Data on female subjects, on subjects at different ages across the life span, or on temporal trajectories of phenotypes after stress that can inform model validity and treatment study design are needed. Overall, preclinical (and clinical) PTSD researchers are increasingly incorporating homologous biological measures to assess markers of risk, response, and treatment outcome. This shift is exciting, as we and many others hope it not only will support translation of drug efficacy from animal models to clinical trials but also will potentially improve predictability of stage II for stage III clinical trials.
Collapse
Affiliation(s)
- Jessica Deslauriers
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, California
| | - Mate Toth
- Department of Behavioural Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andre Der-Avakian
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, California.
| |
Collapse
|
79
|
Cheon SY, Koo BN. Postoperative cognitive dysfunction: advances based on pre-clinical studies. Anesth Pain Med (Seoul) 2018. [DOI: 10.17085/apm.2018.13.2.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- So Yeong Cheon
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Bon-Nyeo Koo
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
80
|
Reijnen A, Geuze E, Eekhout I, Maihofer AX, Nievergelt CM, Baker DG, Vermetten E. Biological profiling of plasma neuropeptide Y in relation to posttraumatic stress symptoms in two combat cohorts. Biol Psychol 2018; 134:72-79. [DOI: 10.1016/j.biopsycho.2018.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 11/02/2017] [Accepted: 02/14/2018] [Indexed: 02/04/2023]
|
81
|
Tillinger A, Horváthová Ľ, Nostramo R, Serova LI, Kvetňanský R, Sabban EL, Mravec B. Glucocorticoid withdrawal affects stress-induced changes of urocortin 2 gene expression in rat adrenal medulla and brain. J Neuroendocrinol 2018; 30:e12595. [PMID: 29604138 DOI: 10.1111/jne.12595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/24/2018] [Indexed: 12/24/2022]
Abstract
Corticotropin-releasing factor is well known activator of the hypothalamic-pituitary-adrenocortical axis, that represents crucial system participating on stress response of the organism. Urocortins are members of the corticotropin-releasing factor family of peptides with proposed effects on neuroendocrine and behavioral stress response mechanisms. Urocortin 2, one of three known urocortins, is present in central and peripheral stress response system and its expression can be augmented by glucocorticoids. In the present study we have examined how glucocorticoid withdrawal affects urocortin 2 gene expression after acute immobilization in the adrenal medulla and selected brain areas in rats. We used pharmacological adrenalectomy to block synthesis of corticosterone. Our results show that the immobilization-induced rise in urocortin 2 mRNA levels in rat adrenal medulla was not inhibited by glucocorticoid withdrawal. On the other hand, observed changes in the brain indicate that the effect of stress and pharmacological adrenalectomy on urocortin 2 gene expression is site-specific. While in the paraventricular nucleus and locus coeruleus the immobilization induced rise of urocortin 2 was not inhibited by pharmacological adrenalectomy in the arcuate nucleus and central amygdala it was. Moreover, we have seen a significant depletion of urocortin 2 plasma levels after immobilization. The immobilization induced rise of urocortin 2 gene expression in rat adrenal medulla and brain areas regulating stress response pathways and preservation of its induction after adrenalectomy suggests a role of urocortin 2 in the neuroendocrine stress response of an organism. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Andrej Tillinger
- Institute of Experimental Endocrinology, Biomedical Research Center Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ľubica Horváthová
- Institute of Experimental Endocrinology, Biomedical Research Center Slovak Academy of Sciences, Bratislava, Slovakia
| | - Regina Nostramo
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA
| | - Richard Kvetňanský
- Institute of Experimental Endocrinology, Biomedical Research Center Slovak Academy of Sciences, Bratislava, Slovakia
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA
| | - Boris Mravec
- Institute of Experimental Endocrinology, Biomedical Research Center Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Physiology, Faculty of Medicine Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
82
|
Camp R, Stier CT, Serova LI, McCloskey J, Edwards JG, Reyes-Zaragoza M, Sabban EL. Cardiovascular responses to intranasal neuropeptide Y in single prolonged stress rodent model of post-traumatic stress disorder. Neuropeptides 2018; 67:87-94. [PMID: 29169656 DOI: 10.1016/j.npep.2017.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/26/2017] [Accepted: 11/08/2017] [Indexed: 01/25/2023]
Abstract
Delivery of neuropeptide Y (NPY) to the brain by intranasal administration shows promise as non-invasive means for preventing or treating PTSD symptoms. Here, radiotelemetry and echocardiography were used to determine effects of intranasal NPY on cardiovascular functions in absence and presence of stress. Male adult Sprague Dawley rats were implanted with radiotelemetric probes, and subjected to single prolonged stress (SPS), followed by intranasal vehicle (V) or NPY (150μg) under conditions shown to prevent development of many of the behavioral neuroendocrine and biochemical impairments. In both groups, mean arterial pressure (MAP) rose rapidly peaking at about 125mmHg, remaining near maximal levels for 1h. SPS also elicited robust rise in heart rate (HR) which was mitigated by intranasal NPY, and significantly lower than V-treated rats 12-50min after exposure to SPS stressors. In the first hr. after SPS, locomotor activity was elevated but only in the V-treated group. By 3h, MAP returned to pre-stress levels in both groups with no further change when monitored for 6days. HR remained elevated during the 6h remaining light phase after SPS. Subsequently HR was at pre-SPS levels during the remaining days. However dark phase HR was low following SPS, gradually recovered over 6days and was associated with reduced activity. When administered in the absence of further stress, intranasal NPY or V elicited similar much smaller, short-lived rises in MAP and HR. Echocardiography revealed no change in HR, stroke volume (SV) or cardiac output (Q) with intranasal NPY in the absence of stress. SPS led to reduced SV and Q but was not affected by NPY. Overall the results demonstrate no major cardiovascular effects of intranasal NPY and indicate possible benefit from transient amelioration of HR response in line with its translational potential to combat PTSD and comorbid impairments.
Collapse
Affiliation(s)
- Robert Camp
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - Charles T Stier
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - Jaclyn McCloskey
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - John G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York 10595, USA
| | - Miguel Reyes-Zaragoza
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York 10595, USA.
| |
Collapse
|
83
|
Jiang JH, Peng YL, Zhang PJ, Xue HX, He Z, Liang XY, Chang M. The ventromedial hypothalamic nucleus plays an important role in anxiolytic-like effect of neuropeptide S. Neuropeptides 2018; 67:36-44. [PMID: 29195839 DOI: 10.1016/j.npep.2017.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 02/08/2023]
Abstract
Neuropeptide S (NPS), the endogenous neuropeptide ligand of NPSR, has been reported to regulate anxiety-related behavior involved in multiple brain regions, including amygdale, locus coeruleus and Barrington's nucleus. However, little research has been conducted on the anxiolytic-like behaviors of NPS on the hypothalamus, which was an important area in defensive behavior. Here, we investigated a role of hypothalamus in anxiolytic-like behaviors of NPS. We found that NPSR protein of mouse distributed mainly in the ventromedial hypothalamus (VMH). And in the single prolonged stress model (SPS), the results showed that NPS mRNA of the mice exposed to SPS was significantly higher than control, while NPSR mRNA was remarkable lower than control in hypothalamus. Further studies found that NPS intra-VMH infusion dose-dependently (1, 10 and 100pmol) induced anxiolytic effects, using elevated plus maze and open field tests. These anxiolytic effects could be blocked by NPSR antagonist (SHA68), but not by picrotoxin (a GABAA receptor antagonist) and sacolfen (a GABAB receptor antagonist). Meanwhile, our data showed that the expression of c-Fos was significantly increased in VMH after NPS delivered into the lateral ventricles. These results cast a new light on the hypothalamic nucleus in the anxiolytic-like effect of NPS system.
Collapse
Affiliation(s)
- Jin Hong Jiang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Ya Li Peng
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Pei Jiang Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Hong Xiang Xue
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Zhen He
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Xue Ya Liang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - M Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
84
|
Sabban EL, Serova LI, Newman E, Aisenberg N, Akirav I. Changes in Gene Expression in the Locus Coeruleus-Amygdala Circuitry in Inhibitory Avoidance PTSD Model. Cell Mol Neurobiol 2018; 38:273-280. [PMID: 28889197 PMCID: PMC11481846 DOI: 10.1007/s10571-017-0548-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/02/2017] [Indexed: 12/28/2022]
Abstract
The locus coeruleus (LC)-amygdala circuit is implicated in playing a key role in responses to emotionally arousing stimuli and in the manifestation of post-traumatic stress disorder (PTSD). Here, we examined changes in gene expression of a number of important mediators of the LC-amygdala circuitry in the inhibition avoidance model of PTSD. After testing for basal acoustic startle response (ASR), rats were exposed to a severe footshock (1.5 mA for 10 s) in the inhibitory avoidance apparatus. They were given contextual situational reminders every 5 day for 25 days. Controls were treated identically but with the footshock inactivated. Animals were re-tested on second ASR and decapitated 1 h later. The shock group had enhanced hyperarousal and several changes in gene expression compared to controls. In the LC, mRNA levels of norepinephrine (NE) biosynthetic enzymes (TH, DBH), NE transporter (NET), NPY receptors (Y1R, Y2R), and CB1 receptor of endocannabinoid system were elevated. In the basolateral amygdala (BLA), there were marked reductions in gene expression for CB1, and especially Y1R, with rise for corticotropin-releasing hormone (CRH) system (CRH, CRH receptor 1), and no significant changes in the central amygdala. Our results suggest a fast forward mechanism in the LC-amygdala circuitry in the shock group.
Collapse
Affiliation(s)
- Esther L Sabban
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY, 10595, USA.
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY, 10595, USA
| | - Elizabeth Newman
- Department of Biochemistry and Molecular Biology, Basic Sciences Building, New York Medical College, Valhalla, NY, 10595, USA
| | - Nurit Aisenberg
- Department of Psychology, University of Haifa, 3498838, Haifa, Israel
| | - Irit Akirav
- Department of Psychology, University of Haifa, 3498838, Haifa, Israel
| |
Collapse
|
85
|
Lisieski MJ, Eagle AL, Conti AC, Liberzon I, Perrine SA. Single-Prolonged Stress: A Review of Two Decades of Progress in a Rodent Model of Post-traumatic Stress Disorder. Front Psychiatry 2018; 9:196. [PMID: 29867615 PMCID: PMC5962709 DOI: 10.3389/fpsyt.2018.00196] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/25/2018] [Indexed: 12/21/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a common, costly, and often debilitating psychiatric condition. However, the biological mechanisms underlying this disease are still largely unknown or poorly understood. Considerable evidence indicates that PTSD results from dysfunction in highly-conserved brain systems involved in stress, anxiety, fear, and reward. Pre-clinical models of traumatic stress exposure are critical in defining the neurobiological mechanisms of PTSD, which will ultimately aid in the development of new treatments for PTSD. Single prolonged stress (SPS) is a pre-clinical model that displays behavioral, molecular, and physiological alterations that recapitulate many of the same alterations observed in PTSD, illustrating its validity and giving it utility as a model for investigating post-traumatic adaptations and pre-trauma risk and protective factors. In this manuscript, we review the present state of research using the SPS model, with the goals of (1) describing the utility of the SPS model as a tool for investigating post-trauma adaptations, (2) relating findings using the SPS model to findings in patients with PTSD, and (3) indicating research gaps and strategies to address them in order to improve our understanding of the pathophysiology of PTSD.
Collapse
Affiliation(s)
- Michael J Lisieski
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Andrew L Eagle
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Alana C Conti
- Research and Development Service, John D. Dingell Veterans Affairs Medical Center, Detroit, MI, United States.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Israel Liberzon
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States.,Mental Health Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
86
|
Lazuko SS, Kuzhel OP, Belyaeva LE, Manukhina EB, Downey HF, Fred Downey H, Tseilikman OB, Komelkova MV, Tseilikman VE. Posttraumatic Stress Disorder Disturbs Coronary Tone and Its Regulatory Mechanisms. Cell Mol Neurobiol 2018; 38:209-217. [PMID: 28676988 PMCID: PMC11481920 DOI: 10.1007/s10571-017-0517-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 06/26/2017] [Indexed: 11/30/2022]
Abstract
Posttraumatic stress disorder (PTSD) is associated with myocardial injury, but changes in coronary regulatory mechanisms in PTSD have not been investigated. This study evaluated the effect of PTSD-inducing stress on coronary tone and its regulation by nitric oxide (NO) and voltage-gated K+ channels. PTSD was induced by exposing rats to predator stress, 15 min daily for 10 days, followed by 14 stress-free days. Presence of PTSD was confirmed by the elevated plus-maze test. Coronary tone was evaluated from changes in coronary perfusion pressure of Langendorff isolated hearts. Predator stress induced significant decreases in coronary tone of isolated hearts and in blood pressure of intact rats. L-NAME, a non-selective NO synthase (NOS) inhibitor, but not S-MT, a selective iNOS inhibitor, and increased coronary tone of control rats. In PTSD rats, both L-NAME and S-MT increased coronary tone. Therefore, the stress-induced coronary vasodilation resulted from NO overproduction by both iNOS and eNOS. NOS induction was apparently due to systemic inflammation as evidenced by increased serum interleukin-1β and C-reactive protein in PTSD rats. Decreased corticosterone in PTSD rats may have contributed to inflammation and its effect on coronary tone. PTSD was also associated with voltage-gated K+ channel dysfunction, which would have also reduced coronary tone.
Collapse
Affiliation(s)
- Svetlana S Lazuko
- Vitebsk State Medical University, Frunze Av 27, Vitebsk, 210023, Republic of Belarus
| | - Olga P Kuzhel
- Vitebsk State Medical University, Frunze Av 27, Vitebsk, 210023, Republic of Belarus
| | - Lyudmila E Belyaeva
- Vitebsk State Medical University, Frunze Av 27, Vitebsk, 210023, Republic of Belarus
| | - Eugenia B Manukhina
- South Ural State University Biomedical School, Lenin Ave, Chelyabinsk, 454080, Russian Federation
- Institute of General Pathology and Pathophysiology, Baltijskaya 8, Moscow, 125315, Russian Federation
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, Texas, 76107, USA
| | | | - H Fred Downey
- Department of Physiology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, Texas, 76107, USA
| | - Olga B Tseilikman
- South Ural State University Biomedical School, Lenin Ave, Chelyabinsk, 454080, Russian Federation
| | - Maria V Komelkova
- South Ural State University Biomedical School, Lenin Ave, Chelyabinsk, 454080, Russian Federation
| | - Vadim E Tseilikman
- South Ural State University Biomedical School, Lenin Ave, Chelyabinsk, 454080, Russian Federation.
| |
Collapse
|
87
|
Sayed S, Van Dam NT, Horn SR, Kautz MM, Parides M, Costi S, Collins KA, Iacoviello B, Iosifescu DV, Mathé AA, Southwick SM, Feder A, Charney DS, Murrough JW. A Randomized Dose-Ranging Study of Neuropeptide Y in Patients with Posttraumatic Stress Disorder. Int J Neuropsychopharmacol 2017; 21:3-11. [PMID: 29186416 PMCID: PMC5795352 DOI: 10.1093/ijnp/pyx109] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/17/2017] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Anxiety and trauma-related disorders are among the most prevalent and disabling medical conditions in the United States, and posttraumatic stress disorder in particular exacts a tremendous public health toll. We examined the tolerability and anxiolytic efficacy of neuropeptide Y administered via an intranasal route in patients with posttraumatic stress disorder. METHODS Twenty-six individuals were randomized in a cross-over, single ascending dose study into 1 of 5 cohorts: 1.4 mg (n=3), 2.8 mg (n=6), 4.6 mg (n=5), 6.8 mg (n=6), and 9.6 mg (n=6). Each individual was dosed with neuropeptide Y or placebo on separate treatment days 1 week apart in random order under double-blind conditions. Assessments were conducted at baseline and following a trauma script symptom provocation procedure subsequent to dosing. Occurrence of adverse events represented the primary tolerability outcome. The difference between treatment conditions on anxiety as measured by the Beck Anxiety Inventory and the State-Trait Anxiety Inventory immediately following the trauma script represented efficacy outcomes. RESULTS Twenty-four individuals completed both treatment days. Neuropeptide Y was well tolerated up to and including the highest dose. There was a significant interaction between treatment and dose; higher doses of neuropeptide Y were associated with a greater treatment effect, favoring neuropeptide Y over placebo on Beck Anxiety Inventory score (F1,20=4.95, P=.038). There was no significant interaction for State-Trait Anxiety Inventory score. CONCLUSIONS Our study suggests that a single dose of neuropeptide Y is well tolerated up to 9.6 mg and may be associated with anxiolytic effects. Future studies exploring the safety and efficacy of neuropeptide Y in stress-related disorders are warranted. The reported study is registered at: http://clinicaltrials.gov (ID: NCT01533519).
Collapse
Affiliation(s)
- Sehrish Sayed
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicholas T Van Dam
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sarah R Horn
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marin M Kautz
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michael Parides
- Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sara Costi
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Katherine A Collins
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian Iacoviello
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York,Click Therapeutics, Inc., New York, New York
| | - Dan V Iosifescu
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York,New York University School of Medicine, New York, New York,Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Aleksander A Mathé
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Steven M Southwick
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut,National Center for PTSD, VA CT Healthcare System, New Haven, Connecticut
| | - Adriana Feder
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dennis S Charney
- Office of the Dean, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James W Murrough
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York,Correspondence: James W. Murrough, MD, Mood and Anxiety Disorders Research Program. Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY 10029 ()
| |
Collapse
|
88
|
Lee B, Shim I, Lee H, Hahm DH. Effect of oleuropein on cognitive deficits and changes in hippocampal brain-derived neurotrophic factor and cytokine expression in a rat model of post-traumatic stress disorder. J Nat Med 2017; 72:44-56. [PMID: 28884427 DOI: 10.1007/s11418-017-1103-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/05/2017] [Indexed: 11/24/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a condition that develops after an individual has experienced a major trauma. This psychopathological response to traumatic stressors induces learning and memory deficits in rats. Oleuropein (OLE), a major compound in olive leaves, has been reported to possess several pharmacological properties, including anti-cancer, anti-diabetic, anti-atherosclerotic and neuroprotective activities. However, the cognitive effects of OLE and its mechanism of action have remained unclear in PTSD. In this study, we examined whether OLE improved spatial cognitive impairment induced in rats following single prolonged stress (SPS), an animal model of PTSD. Male rats were treated intraperitoneally (i.p.) with vehicle or various doses of OLE for 14 consecutive days after the SPS procedure. The SPS procedure resulted in cognitive impairment in the object recognition task and the Morris water maze test, which was reversed by OLE (100 mg/kg, i.p). Additionally, as assessed by immunohistochemistry and reverse transcription-polymerase chain reaction analysis, the administration of OLE significantly alleviated memory-associated decreases in the levels of brain-derived neurotrophic factor and cAMP response element-binding protein and mRNA in the hippocampus. Together, these findings suggest that OLE attenuated SPS-induced cognitive impairment significantly by inhibiting the expression of pro-inflammatory mediators in the rat brain. Thus, OLE reversed several behavioral impairments triggered by the traumatic stress of SPS and might be a potential useful therapeutic intervention for PTSD.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dae-Hyun Hahm
- Acupuncture and Meridian Science Research Center, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
89
|
Aisenberg N, Serova L, Sabban EL, Akirav I. The effects of enhancing endocannabinoid signaling and blocking corticotrophin releasing factor receptor in the amygdala and hippocampus on the consolidation of a stressful event. Eur Neuropsychopharmacol 2017; 27:913-927. [PMID: 28663121 DOI: 10.1016/j.euroneuro.2017.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/20/2017] [Accepted: 06/10/2017] [Indexed: 11/18/2022]
Abstract
Current clinical and pre-clinical data suggest that both cannabinoid agents and blockage of CRF through corticotrophin releasing factor receptor type 1 (CRFr1) may offer therapeutic benefits for post-traumatic stress disorder (PTSD). Here we aim to determine whether they are more effective when combined when microinjected into the basolateral amygdala (BLA) or CA1 area of the hippocampus after exposure to a stressful event in the shock/reminders rat model for PTSD. Injection of the fatty acid amide hydrolase (FAAH) inhibitor URB597 after the shock into either the BLA or CA1 facilitated extinction, and attenuated startle response and anxiety-like behavior. These preventive effects of URB597 were found to be mediated by the CB1 receptor. Intra-BLA and intra-CA1 microinjection of the CRFr1 antagonist, CP-154,526 attenuated startle response. When microinjected into the BLA, CP-154,526 also attenuated freezing behavior during exposure to the first reminder and decreased anxiety-like behavior. The combined treatment of URB597 and CP-154,526 was not more effective than the separate treatments. Finally, mRNA levels of CRF, CRFr1 and CB1r were significantly higher in the BLA of rats exposed to shock and reminders compared to non-shocked rats almost one month after the shock. Taken together, the results show that enhancing endocannabinoid signaling in the amygdala and hippocampus produced a more favorable spectrum of effects than those caused by the CRFr1 antagonist. The findings suggest that FAAH inhibitors may be used as a novel treatment for stress-related anxiety disorders.
Collapse
MESH Headings
- Amidohydrolases/antagonists & inhibitors
- Amidohydrolases/metabolism
- Animals
- Anxiety/drug therapy
- Anxiety/metabolism
- Basolateral Nuclear Complex/drug effects
- Basolateral Nuclear Complex/metabolism
- Benzamides/pharmacology
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/metabolism
- Carbamates/pharmacology
- Disease Models, Animal
- Endocannabinoids/metabolism
- Male
- Memory Consolidation/drug effects
- Memory Consolidation/physiology
- Nootropic Agents/pharmacology
- Pyrimidines/pharmacology
- Pyrroles/pharmacology
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Reflex, Startle/drug effects
- Reflex, Startle/physiology
- Stress Disorders, Post-Traumatic/drug therapy
- Stress Disorders, Post-Traumatic/metabolism
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
Collapse
Affiliation(s)
- Nurit Aisenberg
- Department of Psychology, University of Haifa, Haifa 3498838, Israel
| | - Lidia Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
90
|
Zhang J, Fan Y, Raza MU, Zhan Y, Du XD, Patel PD, Zhu MY. The regulation of corticosteroid receptors in response to chronic social defeat. Neurochem Int 2017; 108:397-409. [DOI: 10.1016/j.neuint.2017.05.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/27/2017] [Accepted: 05/30/2017] [Indexed: 12/24/2022]
|
91
|
Melhorn SJ, Elfers CT, Scott KA, Sakai RR. A closer look at the subordinate population within the visible burrow system. Physiol Behav 2017; 178:110-116. [PMID: 28130085 PMCID: PMC5513744 DOI: 10.1016/j.physbeh.2017.01.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/11/2017] [Accepted: 01/23/2017] [Indexed: 02/05/2023]
Abstract
The visible burrow system (VBS) utilizes the natural social behavior of rodents to model chronic social stress. Classically, when male and female rats are housed together in the VBS a dominance hierarchy rapidly forms with one dominant (DOM) and three subordinate (SUB) males. SUB animals show signs of chronic social stress, including loss of body weight and elevated basal corticosterone. This study furthered examined differences among the SUB population. Quantitative observations across numerous VBS colonies within the Sakai Lab suggest that there is variability in the effects of stress on the SUB population, specifically that some animals may experience more severe effects of chronic social stress than others. To further examine this observation, SUB animals were classified as OMEGA if they received a disproportionate amount of their colonies' wounds. OMEGA animals received more wounds to their body compared to SUB (P<0.0001) and lost significantly more weight throughout the stress period compared to all other VBS-housed animals (group×time interaction P<0.0001). Following VBS housing it was determined the OMGEA also lost lean body mass (P<0.01 vs. controls and DOM), are hyporesponsive to an acute restraint challenge (P<0.01 vs all other groups) and show depressive-like behavior during a forced swim test. Furthermore, expression of neuropeptide Y within the amygdala, known for anxiolytic properties following chronic stress, was elevated among OMEGA (group×region interaction P<0.001). Together these observations suggest that an additional phenotype exists among the SUB animals within a VBS colony and represents the variability of the effects of chronic social stress.
Collapse
Affiliation(s)
- Susan J Melhorn
- Department of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| | - Clinton T Elfers
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Karen A Scott
- Department of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Randall R Sakai
- Department of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
92
|
Endogenously Released Neuropeptide Y Suppresses Hippocampal Short-Term Facilitation and Is Impaired by Stress-Induced Anxiety. J Neurosci 2017; 37:23-37. [PMID: 28053027 DOI: 10.1523/jneurosci.2599-16.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/30/2016] [Accepted: 10/18/2016] [Indexed: 12/15/2022] Open
Abstract
Neuropeptide Y (NPY) has robust anxiolytic properties and is reduced in patients with anxiety disorders. However, the mechanisms by which NPY modulates circuit function to reduce anxiety behavior are not known. Anxiolytic effects of NPY are mediated in the CA1 region of hippocampus, and NPY injection into hippocampus alleviates anxiety symptoms in the predator scent stress model of stress-induced anxiety. The mechanisms that regulate NPY release, and its effects on CA1 synaptic function, are not fully understood. Here we show in acute hippocampal slices from mice that endogenous NPY, released in response to optogenetic stimulation or synaptically evoked spiking of NPY+ cells, suppresses both of the feedforward pathways to CA1. Stimulation of temporoammonic synapses with a physiologically derived spike train causes NPY release that reduces short-term facilitation, whereas the release of NPY that modulates Schaffer collateral synapses requires integration of both the Schaffer collateral and temporoammonic pathways. Pathway specificity of NPY release is conferred by three functionally distinct NPY+ cell types, with differences in intrinsic excitability and short-term plasticity of their inputs. Predator scent stress abolishes the release of endogenous NPY onto temporoammonic synapses, a stress-sensitive pathway, thereby causing enhanced short-term facilitation. Our results demonstrate how stress alters CA1 circuit function through the impairment of endogenous NPY release, potentially contributing to heightened anxiety. SIGNIFICANCE STATEMENT Neuropeptide Y (NPY) has robust anxiolytic properties, and its levels are reduced in patients with post-traumatic stress disorder. The effects of endogenously released NPY during physiologically relevant stimulation, and the impact of stress-induced reductions in NPY on circuit function, are unknown. By demonstrating that NPY release modulates hippocampal synaptic plasticity and is impaired by predator scent stress, our results provide a novel mechanism by which stress-induced anxiety alters circuit function. These studies fill an important gap in knowledge between the molecular and behavioral effects of NPY. This article also advances the understanding of NPY+ cells and the factors that regulate their spiking, which could pave the way for new therapeutic targets to increase endogenous NPY release in patients in a spatially and temporally appropriate manner.
Collapse
|
93
|
Serova L, Mulhall H, Sabban E. NPY1 Receptor Agonist Modulates Development of Depressive-Like Behavior and Gene Expression in Hypothalamus in SPS Rodent PTSD Model. Front Neurosci 2017; 11:203. [PMID: 28469551 PMCID: PMC5395638 DOI: 10.3389/fnins.2017.00203] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/24/2017] [Indexed: 02/04/2023] Open
Abstract
Delivery of neuropeptide Y (NPY) to the brain by intranasal infusion soon after traumatic stress has shown therapeutic potential, and prevented development of many behavioral and neuroendocrine impairments in the single prolonged stress (SPS) animal model of PTSD. Therefore, we examined whether the Y1R preferring agonist [Leu31Pro34]NPY is sufficient to prevent development of SPS induced depressive-like behavioral changes, and hypothalamic gene expression as obtained with intranasal NPY intervention. Male Sprague-Dawely rats were given intranasal infusion of either NPY (150 μg/rat), a low (68 μg /rat), or high (132 μg/rat) dose of [Leu31Pro34]NPY or vehicle immediately following the last SPS stressor, left undisturbed for 1 week and then tested for depressive-like behavior together with naïve unstressed controls. Vehicle treated animals had elevated immobility forced swim test (FST) and reduced sucrose preference, which were not observed in animals given NPY or the higher dose of [Leu31Pro34]NPY. This dose of [Leu31Pro34]NPY, like NPY, also prevented the SPS-elicited induction of CRF mRNA in the mediobasal hypothalamus. However, [Leu31Pro34]NPY did not prevent, but rather enhanced, the SPS-triggered induction of GR and FKBP5 mRNA levels in the mediobasal hypothalamus. Thus, [Leu31Pro34]NPY may be as effective as NPY and displays therapeutic potential for preventing development of depressive-like behaviors and dysregulation of the CRF/HPA system in PTSD. However, due to its different effects compared to NPY on GR and FKBP5 a broader agonist, such as NPY, may be more desirable.
Collapse
Affiliation(s)
- Lidia Serova
- Department of Biochemistry and Molecular Biology, New York Medical CollegeValhalla, NY, USA
| | - Hannah Mulhall
- Department of Biochemistry and Molecular Biology, New York Medical CollegeValhalla, NY, USA
| | - Esther Sabban
- Department of Biochemistry and Molecular Biology, New York Medical CollegeValhalla, NY, USA
| |
Collapse
|
94
|
Effects of moderate treadmill exercise and fluoxetine on behavioural and cognitive deficits, hypothalamic-pituitary-adrenal axis dysfunction and alternations in hippocampal BDNF and mRNA expression of apoptosis – related proteins in a rat model of post-traumatic stress disorder. Neurobiol Learn Mem 2017; 139:165-178. [DOI: 10.1016/j.nlm.2017.01.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 12/14/2016] [Accepted: 01/21/2017] [Indexed: 12/15/2022]
|
95
|
Thorsell A, Mathé AA. Neuropeptide Y in Alcohol Addiction and Affective Disorders. Front Endocrinol (Lausanne) 2017; 8:178. [PMID: 28824541 PMCID: PMC5534438 DOI: 10.3389/fendo.2017.00178] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 07/07/2017] [Indexed: 12/31/2022] Open
Abstract
Neuropeptide Y (NPY), a neuropeptide highly conserved throughout evolution, is present at high levels in the central nervous system (CNS), as well as in peripheral tissues such as the gut and cardiovascular system. The peptide exerts its effects via multiple receptor subtypes, all belonging to the G-protein-coupled receptor superfamily. Of these subtypes, the Y1 and the Y2 are the most thoroughly characterized, followed by the Y5 subtype. NPY and its receptors have been shown to be of importance in central regulation of events underlying, for example, affective disorders, drug/alcohol use disorders, and energy homeostasis. Furthermore, within the CNS, NPY also affects sleep regulation and circadian rhythm, memory function, tissue growth, and plasticity. The potential roles of NPY in the etiology and pathophysiology of mood and anxiety disorders, as well as alcohol use disorders, have been extensively studied. This focus was prompted by early indications for an involvement of NPY in acute responses to stress, and, later, also data pointing to a role in alterations within the CNS during chronic, or repeated, exposure to adverse events. These functions of NPY, in addition to the peptide's regulation of disease states, suggest that modulation of the activity of the NPY system via receptor agonists/antagonists may be a putative treatment mechanism in affective disorders as well as alcohol use disorders. In this review, we present an overview of findings with regard to the NPY system in relation to anxiety and stress, acute as well as chronic; furthermore we discuss post-traumatic stress disorder and, in part depression. In addition, we summarize findings on alcohol use disorders and related behaviors. Finally, we briefly touch upon genetic as well as epigenetic mechanisms that may be of importance for NPY function and regulation. In conclusion, we suggest that modulation of NPY-ergic activity within the CNS, via ligands aimed at different receptor subtypes, may be attractive targets for treatment development for affective disorders, as well as for alcohol use disorders.
Collapse
Affiliation(s)
- Annika Thorsell
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- *Correspondence: Annika Thorsell,
| | - Aleksander A. Mathé
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
96
|
Neuropeptide Y, resilience, and PTSD therapeutics. Neurosci Lett 2016; 649:164-169. [PMID: 27913193 DOI: 10.1016/j.neulet.2016.11.061] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 11/20/2022]
Abstract
Resilience to traumatic stress is a complex psychobiological process that protects individuals from developing posttraumatic stress disorder (PTSD) or other untoward consequences of exposure to extreme stress, including depression. Progress in translational research points toward the neuropeptide Y (NPY) system - among others - as a key mediator of stress response and as a potential therapeutic focus for PTSD. Substantial preclinical evidence supports the role of NPY in the modulation of stress response and in the regulation of anxiety in animal models. Clinical studies testing the safety and efficacy of modulating the NPY system in humans, however, have lagged behind. In the current article, we review the evidence base for targeting the NPY system as a therapeutic approach in PTSD, and consider impediments and potential solutions to therapeutic development.
Collapse
|
97
|
Sun R, Zhang Z, Lei Y, Liu Y, Lu C, Rong H, Sun Y, Zhang W, Ma Z, Gu X. Hippocampal activation of microglia may underlie the shared neurobiology of comorbid posttraumatic stress disorder and chronic pain. Mol Pain 2016; 12:1744806916679166. [PMID: 27852966 PMCID: PMC5117253 DOI: 10.1177/1744806916679166] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/12/2016] [Accepted: 10/17/2016] [Indexed: 12/29/2022] Open
Abstract
The high comorbidity rates of posttraumatic stress disorder and chronic pain have been widely reported, but the underlying mechanisms remain unclear. Emerging evidence suggested that an excess of inflammatory immune activities in the hippocampus involved in the progression of both posttraumatic stress disorder and chronic pain. Considering that microglia are substrates underlying the initiation and propagation of the neuroimmune response, we hypothesized that stress-induced activation of hippocampal microglia may contribute to the pathogenesis of posttraumatic stress disorder-pain comorbidity. We showed that rats exposed to single prolonged stress, an established posttraumatic stress disorder model, exhibited persistent mechanical allodynia and anxiety-like behavior, which were accompanied by increased activation of microglia and secretion of pro-inflammatory cytokines in the hippocampus. Correlation analyses showed that hippocampal activation of microglia was significantly correlated with mechanical allodynia and anxiety-like behavior. Our data also showed that both intraperitoneal and intra-hippocampal injection of minocycline suppressed single prolonged stress-induced microglia activation and inflammatory cytokines accumulation in the hippocampus, and attenuated both single prolonged stress-induced mechanical allodynia and anxiety-like behavior. Taken together, the present study suggests that stress-induced microglia activation in the hippocampus may serve as a critical mechanistic link in the comorbid relationship between posttraumatic stress disorder and chronic pain. The novel concept introduces the possibility of cotreating chronic pain and posttraumatic stress disorder.
Collapse
Affiliation(s)
- Rao Sun
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zuoxia Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yishan Lei
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yue Liu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Cui’e Lu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hui Rong
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yu’e Sun
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wei Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhengliang Ma
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaoping Gu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
98
|
Schmeltzer SN, Herman JP, Sah R. Neuropeptide Y (NPY) and posttraumatic stress disorder (PTSD): A translational update. Exp Neurol 2016; 284:196-210. [PMID: 27377319 PMCID: PMC8375392 DOI: 10.1016/j.expneurol.2016.06.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/12/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a trauma-evoked syndrome, with variable prevalence within the human population due to individual differences in coping and resiliency. In this review, we discuss evidence supporting the relevance of neuropeptide Y (NPY), a stress regulatory transmitter in PTSD. We consolidate findings from preclinical, clinical, and translational studies of NPY that are of relevance to PTSD with an attempt to provide a current update of this area of research. NPY is abundantly expressed in forebrain limbic and brainstem areas that regulate stress and emotional behaviors. Studies in rodents demonstrate a role for NPY in stress responses, anxiety, fear, and autonomic regulation, all relevant to PTSD symptomology. Genetic studies support an association of NPY polymorphisms with stress coping and affect. Importantly, cerebrospinal fluid (CSF) measurements in combat veterans provide direct evidence of NPY association with PTSD diagnosis and symptomology. In addition, NPY involvement in pain, depression, addiction, and metabolism may be relevant to comorbidities associated with PTSD. Collectively, the literature supports the relevance of NPY to PTSD pathophysiology, although knowledge gaps remain. The NPY system is an attractive target in terms of understanding the physiological basis of PTSD as well as treatment of the disorder.
Collapse
Affiliation(s)
- Sarah N Schmeltzer
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Renu Sah
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States; VA Medical Center, Cincinnati, OH, 45220, United States.
| |
Collapse
|
99
|
Kumar JR, Rajkumar R, Jayakody T, Marwari S, Hong JM, Ma S, Gundlach AL, Lai MKP, Dawe GS. Relaxin' the brain: a case for targeting the nucleus incertus network and relaxin-3/RXFP3 system in neuropsychiatric disorders. Br J Pharmacol 2016; 174:1061-1076. [PMID: 27597467 PMCID: PMC5406295 DOI: 10.1111/bph.13564] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022] Open
Abstract
Relaxin‐3 has been proposed to modulate emotional–behavioural functions such as arousal and behavioural activation, appetite regulation, stress responses, anxiety, memory, sleep and circadian rhythm. The nucleus incertus (NI), in the midline tegmentum close to the fourth ventricle, projects widely throughout the brain and is the primary site of relaxin‐3 neurons. Over recent years, a number of preclinical studies have explored the function of the NI and relaxin‐3 signalling, including reports of mRNA or peptide expression changes in the NI in response to behavioural or pharmacological manipulations, effects of lesions or electrical or pharmacological manipulations of the NI, effects of central microinfusions of relaxin‐3 or related agonist or antagonist ligands on physiology and behaviour, and the impact of relaxin‐3 gene deletion or knockdown. Although these individual studies reveal facets of the likely functional relevance of the NI and relaxin‐3 systems for human physiology and behaviour, the differences observed in responses between species (e.g. rat vs. mouse), the clearly identified heterogeneity of NI neurons and procedural differences between laboratories are some of the factors that have prevented a precise understanding of their function. This review aims to draw attention to the current preclinical evidence available that suggests the relevance of the NI/relaxin‐3 system to the pathology and/or symptoms of certain neuropsychiatric disorders and to provide cognizant directions for future research to effectively and efficiently uncover its therapeutic potential. Linked Articles This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc
Collapse
Affiliation(s)
- Jigna Rajesh Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), Singapore.,NUS Graduate School for Integrative Sciences and Engineering (NGS), National University of Singapore, Singapore
| | - Ramamoorthy Rajkumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), Singapore
| | - Tharindunee Jayakody
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), Singapore
| | - Subhi Marwari
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), Singapore
| | - Jia Mei Hong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), Singapore.,NUS Graduate School for Integrative Sciences and Engineering (NGS), National University of Singapore, Singapore
| | - Sherie Ma
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gavin S Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology and Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), Singapore.,NUS Graduate School for Integrative Sciences and Engineering (NGS), National University of Singapore, Singapore
| |
Collapse
|
100
|
Iemolo A, Seiglie M, Blasio A, Cottone P, Sabino V. Pituitary adenylate cyclase-activating polypeptide (PACAP) in the central nucleus of the amygdala induces anxiety via melanocortin receptors. Psychopharmacology (Berl) 2016; 233:3269-77. [PMID: 27376948 PMCID: PMC4982769 DOI: 10.1007/s00213-016-4366-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 06/18/2016] [Indexed: 11/29/2022]
Abstract
RATIONALE Anxiety disorders are the most common mental disorders in the USA. Characterized by feelings of uncontrollable apprehension, they are accompanied by physical, affective, and behavioral symptoms. The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor PAC1 (PAC1R) are highly expressed in the central nucleus of the amygdala (CeA), and they have gained growing attention for their proposed role in mediating the body's response to stress. OBJECTIVES The aim of this study was to evaluate the anxiogenic effects of PACAP in the CeA and its effects on the hypothalamic-pituitary-adrenal (HPA) axis. Furthermore, the mechanism of action of PACAP in the CeA was investigated. METHODS PACAP was microinfused into the CeA of rats, and its effects in the elevated plus maze (EPM), the defensive withdrawal tests, and plasma corticosterone levels were evaluated. The ability of the melanocortin receptor antagonist SHU9119 to block PACAP effect in the EPM was assessed. RESULTS Intra-CeA PACAP exerted a dose-dependent anxiogenic effect and activated the HPA axis. In contrast, PACAP microinfused into the basolateral nucleus of the amygdala (BlA) had no effect. Finally, the anxiogenic effect of intra-CeA PACAP was prevented by SHU9119. CONCLUSIONS These data prove an anxiogenic role for the PACAP system of the CeA and reveal that the melanocortin receptor 4 (MC4R) system of CeA mediates these effects. Our data provide insights into this neuropeptide system as a mechanism for modulating the behavioral and endocrine response to stress and suggest that dysregulations of this system may contribute to the pathophysiology of anxiety-related disorders.
Collapse
Affiliation(s)
- Attilio Iemolo
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, 72 E Concord St, R-612, Boston, MA, 02118, USA
| | - Mariel Seiglie
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, 72 E Concord St, R-612, Boston, MA, 02118, USA
- Graduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Angelo Blasio
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, 72 E Concord St, R-612, Boston, MA, 02118, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, 72 E Concord St, R-612, Boston, MA, 02118, USA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics and Department of Psychiatry, Boston University School of Medicine, 72 E Concord St, R-612, Boston, MA, 02118, USA.
| |
Collapse
|