1
|
da Rocha Zurchimitten G, Oliveira BRF, Maia L, Izídio GS. An enriched environment during adolescence reduced ADHD-related behaviors in SHR and SLA16 male rats. Behav Brain Res 2025:115665. [PMID: 40449738 DOI: 10.1016/j.bbr.2025.115665] [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: 04/02/2025] [Revised: 05/26/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
Environmental enrichment (EE) has emerged as a promising intervention capable of modulating behavior. The isogenic rat strains Spontaneously Hypertensive Rats (SHR) and SHR.LEW-(D4Rat76-D4Mgh11) (SLA16) share an identical genome, differing only in a specific region on chromosome 4. Despite this genetic similarity, these two strains exhibit notable behavioral differences, suggesting a potential influence of environmental and genetic factors. This study aimed to evaluate the effects of continuous EE (40 days, 8 different layouts) during adolescence on the behavior of SHR and SLA16 rat strains. A total of 48 male rats, aged 28 days, were assigned to four experimental groups: SHR-Standard Environment (SHR-SE), SHR-Enriched Environment (SHR-EE), SLA16-Standard Environment (SLA16-SE), and SLA16-Enriched Environment (SLA16-EE) and the following behavioral tests were conducted: Open Field (OF), Marble Burying (MB), Elevated Plus-Maze (EPM), Activity Cage (AC), Novel Object Recognition (NOR), and Plus Maze (PM). Environmental effects were observed in locomotion parameters in the OF, AC, and PM tests (EE < SE); in exploration of aversive areas in the OF and EPM tests (EE < SE); in stereotyped behavior in the MB test (EE < SE); in novelty preference in the NOR and AC tests (EE < SE); and in memory and attention parameters in the PM test (EE > SE). In conclusion, EE demonstrated beneficial effects on the behavior of SHR and SLA16 rat strains and may be considered a potential non-pharmacological intervention strategy for future evaluation in ADHD individuals.
Collapse
Affiliation(s)
| | - Breno Raul Freitas Oliveira
- Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| | - Luara Maia
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| | - Geison Souza Izídio
- Department of Pharmacology - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil; Department of Cellular Biology, Embryology, and Genetics - Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
2
|
Han X, Zhang Y, Petrosky JN, Bald S, Sherva RM, Labadorf A, Cherry JD, Chung J, Farrell K, Abdolmohammadi B, Durape S, Martin BM, Palmisano JN, Farrell JJ, Alvarez VE, Huber BR, Dwyer B, Daneshvar DH, Dams-O'Connor K, Jun GR, Lunetta KL, Goldstein LE, Katz DI, Cantu RC, Shenton ME, Cummings JL, Reiman EM, Stern RA, Alosco ML, Tripodis Y, Farrer LA, Stein TD, Crary JF, McKee AC, Mez J. A structural haplotype in the 17q21.31 MAPT region is associated with increased risk for chronic traumatic encephalopathy endophenotypes. Cell Rep Med 2025; 6:102084. [PMID: 40239644 DOI: 10.1016/j.xcrm.2025.102084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/02/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025]
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative tauopathy associated with repetitive head impact (RHI) exposure. Genetic variation in the 17q21.31 region, containing microtubule-associated protein tau (MAPT), has been implicated in tauopathies but has not been investigated in CTE. The region includes a megabase-long inversion (H1/H2) and copy-number variations, including α, β, and γ segments, which can be characterized as nine segregating structural haplotypes. We leveraged array SNP data and a reference panel across the 17q21.31 region to impute structural haplotypes and test their association with CTE endophenotypes in 447 European ancestry brain donors with RHI exposure. The H1β1γ1 haplotype was significantly associated with dementia and semi-quantitative tau burden in multiple cortical and medial temporal regions commonly affected in CTE. H1β1γ1 differential expression analyses in dorsolateral frontal cortex implicated cis-acting genes and inflammatory pathways. Taken together, the H1β1γ1 haplotype may help explain CTE heterogeneity among those with similar RHI exposure.
Collapse
Affiliation(s)
- Xudong Han
- Bioinformatics Graduate Program, Boston University, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yichi Zhang
- Bioinformatics Graduate Program, Boston University, Boston, MA, USA
| | | | - Sarah Bald
- Bioinformatics Graduate Program, Boston University, Boston, MA, USA
| | - Richard M Sherva
- Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Adam Labadorf
- Bioinformatics Graduate Program, Boston University, Boston, MA, USA; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston VA Healthcare System, Boston, MA, USA
| | - Jonathan D Cherry
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston VA Healthcare System, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jaeyoon Chung
- Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kurt Farrell
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Neuropathology Brain Bank & Research CoRE, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bobak Abdolmohammadi
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Shruti Durape
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA
| | - Brett M Martin
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Joseph N Palmisano
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - John J Farrell
- Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Victor E Alvarez
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston VA Healthcare System, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Bedford VA Healthcare System, Bedford, MA, USA
| | - Bertrand R Huber
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston VA Healthcare System, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Bedford VA Healthcare System, Bedford, MA, USA
| | - Brigid Dwyer
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Daniel H Daneshvar
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Kristen Dams-O'Connor
- Department of Rehabilitation and Human Performance, Brain Injury Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gyungah R Jun
- Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA; Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Lee E Goldstein
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Departments of Biomedical, Electrical & Computer Engineering, Boston University College of Engineering, Boston, MA, USA; Departments of Radiology and Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Douglas I Katz
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Robert C Cantu
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Neurosurgery, Emerson Hospital, Concord, MA, USA
| | - Martha E Shenton
- Boston VA Healthcare System, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, University of Arizona, Arizona State University, Translational Genomics Research Institute, and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Robert A Stern
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Michael L Alosco
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Lindsay A Farrer
- Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA; Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston VA Healthcare System, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Bedford VA Healthcare System, Bedford, MA, USA
| | - John F Crary
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Neuropathology Brain Bank & Research CoRE, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ann C McKee
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston VA Healthcare System, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Bedford VA Healthcare System, Bedford, MA, USA
| | - Jesse Mez
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Chronic Traumatic Encephalopathy Center, Boston University, Boston, MA, USA.
| |
Collapse
|
3
|
Pakkhesal S, Shakouri M, Mosaddeghi-Heris R, Kiani Nasab S, Salehi N, Sharafi A, Ahmadalipour A. Bridging the gap: The endocannabinoid system as a functional fulcrum for benzodiazepines in a novel frontier of anxiety pharmacotherapy. Pharmacol Ther 2025; 267:108799. [PMID: 39862927 DOI: 10.1016/j.pharmthera.2025.108799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/27/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
While benzodiazepines have been a mainstay of the pharmacotherapy of anxiety disorders, their short-term efficacy and risk of abuse have driven the exploration of alternative treatment approaches. The endocannabinoid (eCB) system has emerged as a key modulator of anxiety-related processes, with evidence suggesting dynamic interactions between the eCB system and the GABAergic system, the primary target of benzodiazepines. According to the existing literature, the activation of the cannabinoid receptors has been shown to exert anxiolytic effects, while their blockade or genetic deletion results in heightened anxiety-like responses. Moreover, studies have provided evidence of interactions between the eCB system and benzodiazepines in anxiety modulation. For instance, the attenuation of benzodiazepine-induced anxiolysis by cannabinoid receptor antagonism or genetic variations in the eCB system components in animal studies, have been associated with variations in benzodiazepine response and susceptibility to anxiety disorders. The combined use of cannabinoid-based medications, such as cannabinoid receptor agonists and benzodiazepine co-administration, has shown promise in augmenting anxiolytic effects and reducing benzodiazepine dosage requirements. This article aims to comprehensively review and discuss the current evidence on the involvement of the eCB system as a key modulator of benzodiazepine-related anxiolytic effects, and further, the possible mechanisms by which the region-specific eCB system-GABAergic connectivity modulates the neuro-endocrine/behavioral stress response, providing an inclusive understanding of the complex interplay between the eCB system and benzodiazepines in the context of anxiety regulation, to inform future research and clinical practice.
Collapse
Affiliation(s)
- Sina Pakkhesal
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Shakouri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mosaddeghi-Heris
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Kiani Nasab
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Salehi
- Student Research Committee, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - AmirMohammad Sharafi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Ahmadalipour
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| |
Collapse
|
4
|
Earnhardt-San AL, Baker EC, Cilkiz KZ, Cardoso RC, Ghaffari N, Long CR, Riggs PK, Randel RD, Riley DG, Welsh TH. Evaluation of Prenatal Transportation Stress on DNA Methylation (DNAm) and Gene Expression in the Hypothalamic-Pituitary-Adrenal (HPA) Axis Tissues of Mature Brahman Cows. Genes (Basel) 2025; 16:191. [PMID: 40004522 PMCID: PMC11855312 DOI: 10.3390/genes16020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The experience of prenatal stress results in various physiological disorders due to an alteration of an offspring's methylome and transcriptome. The objective of this study was to determine whether PNS affects DNA methylation (DNAm) and gene expression in the stress axis tissues of mature Brahman cows. Methods: Samples were collected from the paraventricular nucleus (PVN), anterior pituitary (PIT), and adrenal cortex (AC) of 5-year-old Brahman cows that were prenatally exposed to either transportation stress (PNS, n = 6) or were not transported (Control, n = 8). The isolated DNA and RNA samples were, respectively, used for methylation and RNA-Seq analyses. A gene ontology and KEGG pathway enrichment analysis of each data set within each sample tissue was conducted with the DAVID Functional Annotation Tool. Results: The DNAm analysis revealed 3, 64, and 99 hypomethylated and 2, 93, and 90 hypermethylated CpG sites (FDR < 0.15) within the PVN, PIT, and AC, respectively. The RNA-Seq analysis revealed 6, 25, and 5 differentially expressed genes (FDR < 0.15) in the PVN, PIT, and AC, respectively, that were up-regulated in the PNS group relative to the Control group, as well as 24 genes in the PIT that were down-regulated. Based on the enrichment analysis, several developmental and cellular processes, such as maintenance of the actin cytoskeleton, cell motility, signal transduction, neurodevelopment, and synaptic function, were potentially modulated. Conclusions: The methylome and transcriptome were altered in the stress axis tissues of mature cows that had been exposed to prenatal transportation stress. These findings are relevant to understanding how prenatal experiences may affect postnatal neurological functions.
Collapse
Affiliation(s)
- Audrey L. Earnhardt-San
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA
| | - Emilie C. Baker
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Kubra Z. Cilkiz
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Rodolfo C. Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Noushin Ghaffari
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Charles R. Long
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Penny K. Riggs
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Ronald D. Randel
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| |
Collapse
|
5
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
6
|
Mao R, Zhou F, Hong Y, Li Y, Zhu C, Jin L, Li S. CRH upregulates supervillin through ERK and AKT pathways to promote bladder cancer cell migration. Cell Biol Int 2024; 48:1743-1754. [PMID: 39090812 DOI: 10.1002/cbin.12227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Corticotropin-releasing hormone (CRH) has been well documented playing a role in the regulation of cellular processes, immune responses, and inflammatory processes that can influence the occurrence and development of tumors. Supervillin (SVIL) is a membrane-associated and actin-binding protein, which is actively involved in the proliferation, spread, and migration of cancer cells. This work investigated CRH's influence on bladder cancer cells' migration and relevant mechanisms. By using human bladder cancer cells T24 and RT4 in wound healing experiments and transwell assay, we found that the migration ability of the T24 cells was significantly increased after CRH treatment. In vivo experiments showed that CRH significantly promoted the metastases of T24 cells in cell line-derived xenograft (CDX) mouse model. Interestingly, downregulation of SVIL by SVIL-specifc small hairpin RNAs significantly reduced the promoting effect of CRH on bladder cancer cell migration. Furthermore, CRH significantly increased SVIL messenger RNA and protein expression in T24 cells, accompanied with AKT and ERK phosphorylation in T24 cells. Pretreatment with AKT inhibitor (MK2206) blocked the CRH-induced SVIL expression and ERK phosphorylation. Also, inhibition of ERK signaling pathway by U0126 significantly reduced the CRH-induced SVIL expression and AKT phosphorylation. It suggested that cross-talking between AKT and ERK pathways was involved in the effect of CRH on SVIL. Taken together, we demonstrated that CRH induced migration of bladder cancer cells, in which AKT and ERK pathways -SVIL played a key role.
Collapse
Affiliation(s)
- Rongchen Mao
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Feier Zhou
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yali Hong
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yongqi Li
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Chao Zhu
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lai Jin
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Shengnan Li
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Liu L, Huang Z, Zhang J, Wang M, Yue T, Wang W, Wu Y, Zhang Z, Xiong W, Wang C, Wu F, Zhan C, Bi G, Liu J. Hypothalamus-sympathetic-liver axis mediates the early phase of stress-induced hyperglycemia in the male mice. Nat Commun 2024; 15:8632. [PMID: 39366937 PMCID: PMC11452707 DOI: 10.1038/s41467-024-52815-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024] Open
Abstract
Rapid glucose supply is crucial for animal survival during stress response. How the timescale of stress-induced glucose release precisely controlled by hypothalamic corticotropin-releasing hormone (CRH) neurons remains unclear. Here, we show that stress-induced hyperglycemia can be divided into at least two stages in male mice: the first fast stage is mediated by hypothalamus (paraventricular to ventromedial hypothalamus)-sympathetic (raphe pallidus nucleus to intermediolateral nucleus)-liver (HSL) axis activity; the second delayed stage is mediated by adrenal activity. Blocking the activity of HSL axis impairs predatory evoked flight responses, indicating that the HSL pathway activity is necessary for stress coping. We further reveal the intracellular signal cascade for CRH signal in the hypothalamus, which is mediated by GABAA receptor β3 subunit phosphorylation at S408/409, results in prevention of GABAA receptor membrane recruitment. Thus, we uncovered the precise timescale of glucose supply during stress which is mediated by adrenal independent HSL and adrenal dependent pathway respectively.
Collapse
Affiliation(s)
- Ling Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Zhaohuan Huang
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
| | - Jian Zhang
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Mengtian Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Ting Yue
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Wei Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Yue Wu
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Zhi Zhang
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Wei Xiong
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Chao Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Feng Wu
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Cheng Zhan
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Guoqiang Bi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Ji Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
- MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
| |
Collapse
|
8
|
Vandael D, Vints K, Baatsen P, Śliwińska MA, Gabarre S, De Groef L, Moons L, Rybakin V, Gounko NV. Cdk5-dependent rapid formation and stabilization of dendritic spines by corticotropin-releasing factor. Transl Psychiatry 2024; 14:29. [PMID: 38233378 PMCID: PMC10794228 DOI: 10.1038/s41398-024-02749-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/24/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024] Open
Abstract
The neuropeptide corticotropin-releasing factor (CRF) exerts a pivotal role in modulating neuronal activity in the mammalian brain. The effects of CRF exhibit notable variations, depending on factors such as duration of exposure, concentration, and anatomical location. In the CA1 region of the hippocampus, the impact of CRF is dichotomous: chronic exposure to CRF impairs synapse formation and dendritic integrity, whereas brief exposure enhances synapse formation and plasticity. In the current study, we demonstrate long-term effects of acute CRF on the density and stability of mature mushroom spines ex vivo. We establish that both CRF receptors are present in this hippocampal region, and we pinpoint their precise subcellular localization within synapses by electron microscopy. Furthermore, both in vivo and ex vivo data collectively demonstrate that a transient surge of CRF in the CA1 activates the cyclin-dependent kinase 5 (Cdk5)-pathway. This activation leads to a notable augmentation in CRF-dependent spine formation. Overall, these data suggest that upon acute release of CRF in the CA1-SR synapse, both CRF-Rs can be activated and promote synaptic plasticity via activating different downstream signaling pathways, such as the Cdk5-pathway.
Collapse
Affiliation(s)
- Dorien Vandael
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Katlijn Vints
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Pieter Baatsen
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Małgorzata A Śliwińska
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Sergio Gabarre
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Lies De Groef
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Naamsestraat 61 box 2464, 3000, Leuven, Belgium
| | - Lieve Moons
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Naamsestraat 61 box 2464, 3000, Leuven, Belgium
| | - Vasily Rybakin
- National University of Singapore, Department of Microbiology and Immunology, Yng Loo Lin School of Medicine, and Immunology Program, 5 Science Drive 2, Blk MD4, 117545, Singapore, Singapore
| | - Natalia V Gounko
- VIB-KU Leuven Center for Brain & Disease Research, Electron Microscopy Platform & VIB-Bioimaging Core, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium.
- KU Leuven Department of Neurosciences, Leuven Brain Institute, O&N5 Herestraat 49 box 602, 3000, Leuven, Belgium.
| |
Collapse
|
9
|
Kelly EA, Love TM, Fudge JL. Corticotropin-releasing factor-dopamine interactions in male and female macaque: Beyond the classic VTA. Synapse 2024; 78:e22284. [PMID: 37996987 PMCID: PMC10842953 DOI: 10.1002/syn.22284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/25/2023]
Abstract
Dopamine (DA) is involved in stress and stress-related illnesses, including many psychiatric disorders. Corticotropin-releasing factor (CRF) plays a role in stress responses and targets the ventral midbrain DA system, which is composed of DA and non-DA cells, and divided into specific subregions. Although CRF inputs to the midline A10 nuclei ("classic VTA") are known, in monkeys, CRF-containing terminals are also highly enriched in the expanded A10 parabrachial pigmented nucleus (PBP) and in the A8 retrorubral field subregions. We characterized CRF-labeled synaptic terminals on DA (tyrosine hydroxylase, TH+) and non-DA (TH-) cell types in the PBP and A8 regions using immunoreactive electron microscopy (EM) in male and female macaques. CRF labeling was present mostly in axon terminals, which mainly contacted TH-negative dendrites in both subregions. Most CRF-positive terminals had symmetric profiles. In both PBP and A8, CRF symmetric (putative inhibitory) synapses onto TH-negative dendrites were significantly greater than asymmetric (putative excitatory) profiles. This overall pattern was similar in males and females, despite shifts in the size of these effects between regions depending on sex. Because stress and gonadal hormone shifts can influence CRF expression, we also did hormonal assays over a 6-month time period and found little variability in basal cortisol across similarly housed animals at the same age. Together our findings suggest that at baseline, CRF-positive synaptic terminals in the primate PBP and A8 are poised to regulate DA indirectly through synaptic contacts onto non-DA neurons.
Collapse
Affiliation(s)
- E A Kelly
- Departments of Neuroscience, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - T M Love
- Department of Biostatistics, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - J L Fudge
- Departments of Neuroscience, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Psychiatry, Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
10
|
Cichoń L, Pałasz A, Wilczyński KM, Suszka-Świtek A, Żmijowska A, Jelonek I, Janas-Kozik M. Evaluation of Peripheral Blood Concentrations of Phoenixin, Spexin, Nesfatin-1 and Kisspeptin as Potential Biomarkers of Bipolar Disorder in the Pediatric Population. Biomedicines 2023; 12:84. [PMID: 38255190 PMCID: PMC10813295 DOI: 10.3390/biomedicines12010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
There are some initial suggestions in the literature that phoenixin, spexin, nesfatin-1 and kisspeptin may play a role in the pathogenesis of affective disorders. Therefore, they may also be cautiously considered as potential diagnostic or predictive biomarkers of BD. This study aimed to evaluate the levels of the aforementioned neuropeptides in the peripheral blood of children and adolescents with bipolar. This study included 122 individuals: 67 persons with diagnosed bipolar disorder types I and II constituted the study group, and 55 healthy persons were included in the control group. Statistically significant differences in the concentrations of neuropeptides between the control and study groups were noted in relation to nesfatin-1 and spexin (although spexin lost statistical significance after introducing the Bonferroni correction). In a logistic regression analysis, an increased risk of bipolar disorder was noted for a decrease in nesfatin-1 concentration. Lower levels of nesfatin-1 seemed to be a significant risk factor for the development of bipolar disorder types I and II. Furthermore, the occurrence of bipolar disorder was associated with significantly elevated levels of spexin. None of the analyzed neuropeptides was significantly correlated with the number of symptoms of bipolar disorder.
Collapse
Affiliation(s)
- Lena Cichoń
- Department of Developmental Age Psychiatry and Psychotherapy, John Paul II Pediatric Centre in Sosnowiec, Faculty of Medical Sciences, Medical University of Silesia in Katowice, ul. Zapolskiej 3, 41-218 Sosnowiec, Poland
| | - Artur Pałasz
- Department of Histology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, ul. Medyków 18, 40-752 Katowice, Poland
| | - Krzysztof M. Wilczyński
- Department of Developmental Age Psychiatry and Psychotherapy, John Paul II Pediatric Centre in Sosnowiec, Faculty of Medical Sciences, Medical University of Silesia in Katowice, ul. Zapolskiej 3, 41-218 Sosnowiec, Poland
| | - Aleksandra Suszka-Świtek
- Department of Histology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, ul. Medyków 18, 40-752 Katowice, Poland
| | - Anna Żmijowska
- Department of Developmental Age Psychiatry and Psychotherapy, John Paul II Pediatric Centre in Sosnowiec, Faculty of Medical Sciences, Medical University of Silesia in Katowice, ul. Zapolskiej 3, 41-218 Sosnowiec, Poland
| | - Ireneusz Jelonek
- Department of Developmental Age Psychiatry and Psychotherapy, John Paul II Pediatric Centre in Sosnowiec, Faculty of Medical Sciences, Medical University of Silesia in Katowice, ul. Zapolskiej 3, 41-218 Sosnowiec, Poland
| | - Małgorzata Janas-Kozik
- Department of Developmental Age Psychiatry and Psychotherapy, John Paul II Pediatric Centre in Sosnowiec, Faculty of Medical Sciences, Medical University of Silesia in Katowice, ul. Zapolskiej 3, 41-218 Sosnowiec, Poland
| |
Collapse
|
11
|
Rayatpour A, Radahmadi M, Izadi MS, Ghasemi M. Effects of sub-chronic CRH administration into the hypothalamic paraventricular and central amygdala nuclei in male rats with a focus on food intake biomarkers. AN ACAD BRAS CIENC 2023; 95:e20200221. [PMID: 38088701 DOI: 10.1590/0001-3765202320200221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/22/2020] [Indexed: 12/18/2023] Open
Abstract
CRH neurons are found in the paraventricular nucleus(PVN) and central amygdala(CeA) nuclei. This study investigated the effects of sub-chronic CRH administration into the PVN and CeA nuclei on food intake biomarkers in rats divided into five groups: control, two shams, and two CRH-PVN and CRH-CeA groups(receiving CRH in nuclei for seven days). The CRH-PVN group had significantly higher cumulative food intake and food intake trends than the CRH-CeA group. The CRH-CeA and CRH-PVN groups exhibited significant increases in food intake during hours 1 and 2, respectively. Moreover, to be time-dependent, food intake is modulated by different brain nuclei. The CRH signaling pathway appeared to be activated later in the PVN than CeA. Both groups exhibited significantly higher leptin levels, the CRH-PVN group exhibited higher ghrelin levels and lower glucose levels. Repetitive administration of CRH into the PVN and CeA significantly reduced body weight differences. CRH administration into the PVN affected both leptin and ghrelin levels, but ghrelin had a greater impact on glucose variations and cumulative food intake than leptin. Finally, CRH administration into the PVN and CeA likely activated the HPA axis, and the CeA had a greater impact on the stress circuit than on food intake behavior.
Collapse
Affiliation(s)
- Atefeh Rayatpour
- Isfahan University of Medical Sciences, Department of Physiology, School of Medicine, Hezar Jerib street, Isfahan, Iran
| | - Maryam Radahmadi
- Isfahan University of Medical Sciences, Department of Physiology, School of Medicine, Hezar Jerib street, Isfahan, Iran
| | - Mina S Izadi
- Isfahan University of Medical Sciences, Department of Physiology, School of Medicine, Hezar Jerib street, Isfahan, Iran
| | - Maedeh Ghasemi
- Isfahan University of Medical Sciences, Department of Physiology, School of Medicine, Hezar Jerib street, Isfahan, Iran
| |
Collapse
|
12
|
Jovanovic P, Pool AH, Morones N, Wang Y, Novinbakht E, Keshishian N, Jang K, Oka Y, Riera CE. A sex-specific thermogenic neurocircuit induced by predator smell recruiting cholecystokinin neurons in the dorsomedial hypothalamus. Nat Commun 2023; 14:4937. [PMID: 37582805 PMCID: PMC10427624 DOI: 10.1038/s41467-023-40484-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 07/31/2023] [Indexed: 08/17/2023] Open
Abstract
Olfactory cues are vital for prey animals like rodents to perceive and evade predators. Stress-induced hyperthermia, via brown adipose tissue (BAT) thermogenesis, boosts physical performance and facilitates escape. However, many aspects of this response, including thermogenic control and sex-specific effects, remain enigmatic. Our study unveils that the predator odor trimethylthiazoline (TMT) elicits BAT thermogenesis, suppresses feeding, and drives glucocorticoid release in female mice. Chemogenetic stimulation of olfactory bulb (OB) mitral cells recapitulates the thermogenic output of this response and associated stress hormone corticosterone release in female mice. Neuronal projections from OB to medial amygdala (MeA) and dorsomedial hypothalamus (DMH) exhibit female-specific cFos activity toward odors. Cell sorting and single-cell RNA-sequencing of DMH identify cholecystokinin (CCK)-expressing neurons as recipients of predator odor cues. Chemogenetic manipulation and neuronal silencing of DMHCCK neurons further implicate these neurons in the propagation of predator odor-associated thermogenesis and food intake suppression, highlighting their role in female stress-induced hyperthermia.
Collapse
Affiliation(s)
- Predrag Jovanovic
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Allan-Hermann Pool
- Department of Neuroscience, Department of Anesthesiology and Pain Management, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nancy Morones
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Yidan Wang
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Edward Novinbakht
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Nareg Keshishian
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Kaitlyn Jang
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Celine E Riera
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
- Department of Neurology, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, CA, 90048, USA.
| |
Collapse
|
13
|
Raise-Abdullahi P, Meamar M, Vafaei AA, Alizadeh M, Dadkhah M, Shafia S, Ghalandari-Shamami M, Naderian R, Afshin Samaei S, Rashidy-Pour A. Hypothalamus and Post-Traumatic Stress Disorder: A Review. Brain Sci 2023; 13:1010. [PMID: 37508942 PMCID: PMC10377115 DOI: 10.3390/brainsci13071010] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Humans have lived in a dynamic environment fraught with potential dangers for thousands of years. While fear and stress were crucial for the survival of our ancestors, today, they are mostly considered harmful factors, threatening both our physical and mental health. Trauma is a highly stressful, often life-threatening event or a series of events, such as sexual assault, war, natural disasters, burns, and car accidents. Trauma can cause pathological metaplasticity, leading to long-lasting behavioral changes and impairing an individual's ability to cope with future challenges. If an individual is vulnerable, a tremendously traumatic event may result in post-traumatic stress disorder (PTSD). The hypothalamus is critical in initiating hormonal responses to stressful stimuli via the hypothalamic-pituitary-adrenal (HPA) axis. Linked to the prefrontal cortex and limbic structures, especially the amygdala and hippocampus, the hypothalamus acts as a central hub, integrating physiological aspects of the stress response. Consequently, the hypothalamic functions have been attributed to the pathophysiology of PTSD. However, apart from the well-known role of the HPA axis, the hypothalamus may also play different roles in the development of PTSD through other pathways, including the hypothalamic-pituitary-thyroid (HPT) and hypothalamic-pituitary-gonadal (HPG) axes, as well as by secreting growth hormone, prolactin, dopamine, and oxytocin. This review aims to summarize the current evidence regarding the neuroendocrine functions of the hypothalamus, which are correlated with the development of PTSD. A better understanding of the role of the hypothalamus in PTSD could help develop better treatments for this debilitating condition.
Collapse
Affiliation(s)
| | - Morvarid Meamar
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
- Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Alizadeh
- Department of Basic Medical Sciences, Faculty of Medicine, Qom Medical Sciences, Islamic Azad University, Qom, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sakineh Shafia
- Immunogenetics Research Center, Department of Physiology, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Ramtin Naderian
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed Afshin Samaei
- Department of Neurology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
- Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
14
|
Fudge JL, Kelly EA, Hackett TA. Corticotropin Releasing Factor (CRF) Coexpression in GABAergic, Glutamatergic, and GABA/Glutamatergic Subpopulations in the Central Extended Amygdala and Ventral Pallidum of Young Male Primates. J Neurosci 2022; 42:8997-9010. [PMID: 36280261 PMCID: PMC9732834 DOI: 10.1523/jneurosci.1453-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
The central extended amygdala (CEA) and ventral pallidum (VP) are involved in diverse motivated behaviors based on rodent models. These structures are conserved, but expanded, in higher primates, including human. Corticotropin releasing factor (CRF), a canonical "stress molecule" associated with the CEA and VP circuitry across species, is dynamically regulated by stress and drugs of abuse and misuse. CRF's effects on circuits critically depend on its colocation with primary "fast" transmitters, making this crucial for understanding circuit effects. We surveyed the distribution and colocalization of CRF-, VGluT2- (vesicular glutamate transporter 2), and VGAT- (vesicular GABA transporter) mRNA in specific subregions of the CEA and VP in young male monkeys. Although CRF-containing neurons were clustered in the lateral central bed nucleus (BSTLcn), the majority were broadly dispersed throughout other CEA subregions, and the VP. CRF/VGAT-only neurons were highest in the BSTLcn, lateral central amygdala nucleus (CeLcn), and medial central amygdala nucleus (CeM) (74%, 73%, and 85%, respectively). In contrast, lower percentages of CRF/VGAT only neurons populated the sublenticular extended amygdala (SLEAc), ventrolateral bed nucleus (BSTLP), and VP (53%, 54%, 17%, respectively), which had higher complements of CRF/VGAT/VGluT2-labeled neurons (33%, 29%, 67%, respectively). Thus, the majority of CRF-neurons at the "poles" (BSTLcn and CeLcn/CeM) of the CEA are inhibitory, while the "extended" BSTLP and SLEAc subregions, and neighboring VP, have a more complex profile with admixtures of "multiplexed" excitatory CRF neurons. CRF's colocalization with its various fast transmitters is likely circuit-specific, and relevant for understanding CRF actions on specific target sites.SIGNIFICANCE STATEMENT The central extended amygdala (CEA) and ventral pallidum (VP) regulate multiple motivated behaviors through differential downstream projections. The stress neuropeptide corticotropin releasing factor (CRF) is enriched in the CEA, and is thought to "set the gain" through modulatory effects on coexpressed primary transmitters. Using protein and transcript assays in monkey, we found that CRF neurons are broadly and diffusely distributed in CEA and VP. CRF mRNA+ neurons colocalize with VGAT (GABA) and VGluT2 (glutamate) mRNAs in different proportions depending on subregion. CRF mRNA was also coexpressed in a subpopulation of VGAT/VGluT2 mRNA ("multiplexed") cells, which were most prominent in the VP and "pallidal"-like parts of the CEA. Heterogeneous CRF and fast transmitter coexpression across CEA/VP subregions implies circuit-specific effects.
Collapse
Affiliation(s)
- Julie L Fudge
- Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642
| | - Emily A Kelly
- Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642
| | - Troy A Hackett
- Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
15
|
Kupcova I, Danisovic L, Grgac I, Harsanyi S. Anxiety and Depression: What Do We Know of Neuropeptides? Behav Sci (Basel) 2022; 12:262. [PMID: 36004833 PMCID: PMC9405013 DOI: 10.3390/bs12080262] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
In modern society, there has been a rising trend of depression and anxiety. This trend heavily impacts the population's mental health and thus contributes significantly to morbidity and, in the worst case, to suicides. Modern medicine, with many antidepressants and anxiolytics at hand, is still unable to achieve remission in many patients. The pathophysiology of depression and anxiety is still only marginally understood, which encouraged researchers to focus on neuropeptides, as they are a vast group of signaling molecules in the nervous system. Neuropeptides are involved in the regulation of many physiological functions. Some act as neuromodulators and are often co-released with neurotransmitters that allow for reciprocal communication between the brain and the body. Most studied in the past were the antidepressant and anxiolytic effects of oxytocin, vasopressin or neuropeptide Y and S, or Substance P. However, in recent years, more and more novel neuropeptides have been added to the list, with implications for the research and development of new targets, diagnostic elements, and even therapies to treat anxiety and depressive disorders. In this review, we take a close look at all currently studied neuropeptides, their related pathways, their roles in stress adaptation, and the etiology of anxiety and depression in humans and animal models. We will focus on the latest research and information regarding these associated neuropeptides and thus picture their potential uses in the future.
Collapse
Affiliation(s)
- Ida Kupcova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| | - Ivan Grgac
- Institute of Anatomy, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (I.K.); (L.D.)
| |
Collapse
|
16
|
Zhou L, Wang T, Yu Y, Li M, Sun X, Song W, Wang Y, Zhang C, Fu F. The etiology of poststroke-depression: a hypothesis involving HPA axis. Biomed Pharmacother 2022; 151:113146. [PMID: 35643064 DOI: 10.1016/j.biopha.2022.113146] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/06/2022] [Accepted: 05/15/2022] [Indexed: 11/30/2022] Open
Abstract
Approximately, one in three ischemic stroke survivors suffered from depression, namely, post-stroke depression (PSD). PSD affects functional rehabilitation and may lead to poor quality of life of patients. There are numerous explanations about the etiologies of PSD. Here, we speculated that PSD are likely to be the result of specific changes in brain pathology. We hypothesized that the stroke-induced hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis plays an important role in PSD. Stroke initiates a complex sequence of events in neuroendocrine system including HPA axis. The HPA axis is involved in the pathophysiology of depression, especially, the overactivity of the HPA axis occurs in major depressive disorder. This review summarizes the possible etiologies of PSD, focusing on the stroke-induced activation of HPA axis, mainly including the stress followed by severe brain damage and the proinflammatory cytokines release. The role of hyperactive of HPA axis in PSD was discussed in detail, which includes the role of high level corticotropin-releasing hormone in PSD, the effects of glucocorticoids on the alterations in specific brain structures, the expression of enzymes, excitotoxicity, the change in intestinal permeability, and the activation of microglia. The relationship between neuroendocrine regulation and inflammation was also described. Finally, the therapy of PSD by regulating HPA axis, neuroendocrine, and immunity was discussed briefly. Nevertheless, the change of HPA axis and the occurring of PSD maybe interact and promote on each other, and future investigations should explore this hypothesis in more depth.
Collapse
Affiliation(s)
- Lin Zhou
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Yawen Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Mingan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Xiaohui Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Wenhao Song
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Yunjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Ce Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| |
Collapse
|
17
|
David M, Serena B, Jeremy B, Madeline T, Bernard BW. CRF-receptor1 modulation of the dopamine projection to prelimbic cortex facilitates cognitive flexibility after acute and chronic stress. Neurobiol Stress 2022; 16:100424. [PMID: 35005102 PMCID: PMC8718497 DOI: 10.1016/j.ynstr.2021.100424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/07/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022] Open
Abstract
Stress reduces cognitive flexibility and dopamine D1 receptor-related activity in the prelimbic cortex (PL), effects hypothesized to depend on reduced corticotropic releasing factor receptor type 1 (CRFr1) regulation of dopamine neurons in the ventral tegmental area (VTA). We assessed this hypothesis in rats by examining the effect of chronic unpredictable restraint stress (CUS), mild acute stress, or their combination on cognitive flexibility, CRFr1 expression in the VTA and D1-related activity in PL. In Experiment 1, rats received either CUS or equivalent handling for 14 days before being trained to press two levers to earn distinct food outcomes. Initial learning was assessed using an outcome devaluation test after which cognitive flexibility was assessed by reversing the outcomes earned by the actions. Prior to each reversal training session, half the CUS and controls receiving acute stress with action-outcome updating assessed using a second devaluation test and CRFr1 expression in the VTA assessed using in-situ hybridisation. Although CUS did not itself affect action-outcome learning, its combination with acute stress blocked reversal learning and decreased VTA CRFr1 expression after acute shock. The relationship between these latter two effects was assessed in Experiment 2 by pharmacologically disconnecting the VTA and PL, unilaterally blocking neurons expressing CRFr1 in the VTA and D1 receptors in the contralateral PL during reversal learning after acute stress. Acute stress again blocked reversal learning but only in the group with VTA-PL disconnection, demonstrating that VTA CRFr1-induced facilitation of dopaminergic activity in the PL is necessary for maintaining cognitive flexibility after acute stress. [250]. Acute stress increased CRF receptor1 expression in the VTA. Chronic stress attenuated the effect of acute stress on CRFr1 expression. Chronic stress plus acute stress produced a loss of cognitive flexibility. Blocking VTA CFRr1 and dopamine D1r in PL reduced cognitive flexibility following stress.
Collapse
Affiliation(s)
- Mor David
- School of Medical Sciences, University of Sydney, Australia
| | - Becchi Serena
- Decision Neuroscience Lab, University of New South Wales, Australia
| | - Bowring Jeremy
- School of Medical Sciences, University of Sydney, Australia
| | | | | |
Collapse
|
18
|
Wang Y, Zhang Y, Shi Z, Di T, Yu W, Chen L. Exposure of male mice to perfluorooctanoic acid induces anxiety-like behaviors by increasing corticotropin-releasing factor in the basolateral amygdala complex. CHEMOSPHERE 2022; 287:132170. [PMID: 34826932 DOI: 10.1016/j.chemosphere.2021.132170] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 06/13/2023]
Abstract
Perfluorooctanoic acid (PFOA), a hazardous environmental pollutant, has been found to enhance hepatic synthesis of fibroblast growth factor 21 (FGF21). FGF21 can enter the brain and increase the expression of corticotropin-releasing factor (CRF) in the paraventricular nucleus (PVN). In this study, adult male mice were orally administered PFOA to evaluate how it regulates emotion. Exposure of mice to PFOA (1 mg kg-1 bw) for 10 consecutive days (PFOA-mice) caused anxiety-like behaviors and a peroxisome proliferator-activated receptor α (PPARα)-dependent increase in hepatic FGF21 synthesis. The levels of CRF expression in not only PVN but also basolateral amygdala complex (BLA) neurons of PFOA-mice were increased via FGF receptor 1 (FGF-R1) activation. However, the microinjection of FGF-R1 or CRF 1 receptor (CRF-R1) antagonist in the BLA rather than the PVN of PFOA-mice could relieve their anxiety-like behaviors. In addition, external capsule-BLA synaptic transmission in PFOA-mice was enhanced by increasing CRF-R1-mediated presynaptic glutamate release, which was corrected by the blockade of PPARα, FGF-R1 and CRF-R1 or the inhibition of PKA. Furthermore, the threshold of frequency-dependent long-term potentiation (LTP) induction was decreased in the BLA of PFOA-mice, which depended on the activation of PPARα, FGF-R1, CRF-R1, PKA and NMDA receptor (NMDAR), whereas long-term depression (LTD) induction was unchanged. Thus, the results indicate that the exposure of male mice to PFOA (1 mg kg-1 bw) enhances CRF expression in BLA neurons by increasing hepatic FGF21 synthesis, which then enhances CRF-R1-mediated presynaptic glutamate release to facilitate NMDAR-dependent BLA-LTP induction, leading to the production of anxiety-like behaviors.
Collapse
Affiliation(s)
- Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Yajie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Zhaochun Shi
- Department of Neurology, First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Tingting Di
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Wenfeng Yu
- Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, 550004, China.
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
19
|
Sukhareva EV. The role of the corticotropin-releasing hormone and its receptors in the regulation of stress response. Vavilovskii Zhurnal Genet Selektsii 2021; 25:216-223. [PMID: 34901719 PMCID: PMC8627883 DOI: 10.18699/vj21.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/19/2020] [Accepted: 10/06/2020] [Indexed: 11/19/2022] Open
Abstract
Stress is an essential part of everyday life. The neuropeptide corticotropin-releasing hormone (CRH, also
called CRF and corticoliberin) plays a key role in the integration of neuroendocrine, autonomic and behavioral
responses to stress. The activation of the hypothalamic-pituitary-adrenal axis (HPA axis) by neurons of the paraventricular hypothalamic nucleus (PVN), the primary site of synthesis CRH, triggers stress reactions. In addition to the
hypothalamus, CRH is widespread in extrahypothalamic brain structures, where it functions as a neuromodulator
for coordination and interaction between the humoral and behavioral aspects of a stress response. The axons of
neurons expressing CRH are directed to various structures of the brain, where the neuropeptide interacts with
specific receptors (CRHR1, CRHR2) and can affect various mediator systems that work together to transmit signals
to different brain regions to cause many reactions to stress. Moreover, the effect of stress on brain functions varies
from behavioral adaptation to increased survival and increased risk of developing mental disorders. Disturbances
of the CRH system regulation are directly related to such disorders: mental pathologies (depression, anxiety, addictions), deviations of neuroendocrinological functions, inflammation, as well as the onset and development of
neurodegenerative diseases such as Alzheimer’s disease. In addition, the role of CRH as a regulator of the neurons
structure in the areas of the developing and mature brain has been established. To date, studies have been conducted in which CRHR1 is a target for antidepressants, which are, in fact, antagonists of this receptor. In this regard,
the study of the participation of the CRH system and its receptors in negative effects on hormone-dependent
systems, as well as the possibility of preventing them, is a promising task of modern physiological genetics. In this
review, attention will be paid to the role of CRH in the regulation of response to stress, as well as to the involvement
of extrahypothalamic CRH in pathophysiology and the correction of mental disorders.
Collapse
Affiliation(s)
- E V Sukhareva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
20
|
Malta MB, Martins J, Novaes LS, Dos Santos NB, Sita L, Camarini R, Scavone C, Bittencourt J, Munhoz CD. Norepinephrine and Glucocorticoids Modulate Chronic Unpredictable Stress-Induced Increase in the Type 2 CRF and Glucocorticoid Receptors in Brain Structures Related to the HPA Axis Activation. Mol Neurobiol 2021; 58:4871-4885. [PMID: 34213722 DOI: 10.1007/s12035-021-02470-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/22/2021] [Indexed: 12/26/2022]
Abstract
The stress response is multifactorial and enrolls circuitries to build a coordinated reaction, leading to behavioral, endocrine, and autonomic changes. These changes are mainly related to the hypothalamus-pituitary-adrenal (HPA) axis activation and the organism's integrity. However, when self-regulation is ineffective, stress becomes harmful and predisposes the organism to pathologies. The chronic unpredictable stress (CUS) is a widely used experimental model since it induces physiological and behavioral changes and better mimics the stressors variability encountered in daily life. Corticotropin-releasing factor (CRF) and glucocorticoids (GCs) are deeply implicated in the CUS-induced physiological and behavioral changes. Nonetheless, the CUS modulation of CRF receptors and GR and the norepinephrine role in extra-hypothalamic brain areas were not well explored. Here, we show that 14 days of CUS induced a long-lasting HPA axis hyperactivity evidenced by plasmatic corticosterone increase and adrenal gland hypertrophy, which was dependent on both GCs and NE release induced by each stress session. CUS also increased CRF2 mRNA expression and GR protein levels in fundamental brain structures related to HPA regulation and behavior, such as the lateral septal nucleus intermedia part (LSI), ventromedial hypothalamic nucleus (VMH), and central nucleus of the amygdala (CeA). We also showed that NE participates in the CUS-induced increase in CRF2 and GR levels in the LSI, reinforcing the locus coeruleus (LC) involvement in the HPA axis modulation. Despite the CUS-induced molecular changes in essential areas related to anxiety-like behavior, this phenotype was not observed in CUS animals 24 h after the last stress session.
Collapse
Affiliation(s)
- Marilia B Malta
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Joelcimar Martins
- Central of Facilities, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, 05508-000, Brazil
| | - Leonardo S Novaes
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Nilton B Dos Santos
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Luciane Sita
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, 05508-000, Brazil
| | - Rosana Camarini
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil
| | - Jackson Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, 05508-000, Brazil.,Center for Neurosciences and Behavior, Institute of Psychology, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Carolina D Munhoz
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Av. Prof. Lineu Prestes, 1524, room 323, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
21
|
Zheng Y, Zhang YM, Tang ZS, Du JK, Guo DW, Xu YJ, Sheng H, Lu JQ, Ni X. Spatial learning and memory deficits induced by prenatal glucocorticoid exposure depend on hippocampal CRHR1 and CXCL5 signaling in rats. J Neuroinflammation 2021; 18:85. [PMID: 33810797 PMCID: PMC8019183 DOI: 10.1186/s12974-021-02129-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background Prenatal synthetic glucocorticoid (sGC) exposure increases the susceptibility to cognitive and affective disorders in postnatal life. We previously demonstrated that prenatal sGC exposure results in an increase in corticotropin-releasing hormone (CRH) receptor type 1 (CRHR1) expression in the hippocampus of rats, and CRHR1 is involved in synapse formation via regulation of C-X-C chemokine ligand 5 (CXCL5) in hippocampus. We sought to investigate that the roles of CRHR1 and CXCL5 in learning and memory impairment caused by prenatal sGC exposure. Methods Pregnant rats were administered with saline or dexamethasone (DEX) from gestational day (GD) 14 to GD21. DEX offspring at 2-day old were treated with saline and CRHR1 antagonists (antalarmin and CP154526) for 7 days. Some DEX offspring received intra-hippocampal injection of AAV9 carrying CXCL5 gene. Spatial learning and memory was assessed by Morris water maze test. Immunofluorescence analysis was applied to show synapsin I and PSD95 signals in hippocampus. Synapsin I and PSD95 protein level and CXCL5 concentration were determined by western blotting and ELISA, respectively. Organotypic hippocampal slice cultures were used to investigate the effect of DEX on CXCL5 production in vitro. Results Both male and female DEX offspring displayed impairment of spatial learning and memory in adulthood. Synapsin I and PSD95 signals and CXCL5 levels were decreased in DEX offspring. DEX offspring with antalarmin and CP154526 treatment showed improved spatial learning and memory. Antalarmin and CP154526 treatment increased synapsin I and PSD95 signals and CXCL5 concentration in hippocampus. Bilaterally hippocampal injection of AAV9 carrying CXCL5 gene improved the spatial learning and memory and increased CXCL5 concentration and synapsin I and PSD95 levels in hippocampus. DEX dose-dependently suppressed CXCL5 production in cultured hippocammpal slices, which was prevented by antalarmin treatment. Conclusion CRHR1 and CXCL5 signaling in the hippocampus are involved in spatial learning and memory deficits caused by prenatal DEX exposure. CRHR1 activation contributes to decreased CXCL5 production in hippocampus induced by prenatal DEX treatment. Our study provides a molecular basis of prenatal GC exposure programming spatial learning and memory.
Collapse
Affiliation(s)
- You Zheng
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China.,Department of Physiology, Navy Medical University, Shanghai, 200433, China.,Laboratory of Cell Engineering, Institute of Biotechnology, Beijing, 100071, China
| | - Yan-Min Zhang
- Department of Physiology, Navy Medical University, Shanghai, 200433, China.,School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Zheng-Shan Tang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Jian-Kui Du
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China
| | - De-Wei Guo
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Yong-Jun Xu
- Department of Physiology, Navy Medical University, Shanghai, 200433, China
| | - Hui Sheng
- Department of Physiology, Navy Medical University, Shanghai, 200433, China
| | - Jian-Qiang Lu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Xin Ni
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, 410008, China. .,Department of Physiology, Navy Medical University, Shanghai, 200433, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China.
| |
Collapse
|
22
|
Silberstein S, Liberman AC, Dos Santos Claro PA, Ugo MB, Deussing JM, Arzt E. Stress-Related Brain Neuroinflammation Impact in Depression: Role of the Corticotropin-Releasing Hormone System and P2X7 Receptor. Neuroimmunomodulation 2021; 28:52-60. [PMID: 33845478 DOI: 10.1159/000515130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/07/2021] [Indexed: 11/19/2022] Open
Abstract
Depression and other psychiatric stress-related disorders are leading causes of disability worldwide. Up to date, treatments of mood disorders have limited success, most likely due to the multifactorial etiology of these conditions. Alterations in inflammatory processes have been identified as possible pathophysiological mechanisms in psychiatric conditions. Here, we review the main features of 2 systems involved in the control of these inflammatory pathways: the CRH system as a key regulator of the stress response and the ATP-gated ion-channel P2X7 receptor (P2X7R) involved in the control of immune functions. The pathophysiology of depression as a stress-related psychiatric disorder is depicted in terms of the impact of CRH and P2X7R function on inflammatory pathways in the brain. Understanding pathogenesis of affective disorders will lead to the development of therapies for treatment of depression and other stress-related diseases.
Collapse
Affiliation(s)
- Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Ana Clara Liberman
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Paula Ayelén Dos Santos Claro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Maria Belén Ugo
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | | | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- DFBMC, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
23
|
Savareh BA, Bashiri A, Hatef MM, Hatef B. Prediction of salivary cortisol level by electroencephalography features. ACTA ACUST UNITED AC 2020; 66:275-284. [PMID: 34062630 DOI: 10.1515/bmt-2020-0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 11/23/2020] [Indexed: 11/15/2022]
Abstract
Change in cortisol affects brain EEG signals. So, the identification of the significant EEG features which are sensitized to cortisol concentration was the aim of the present study. From 468 participated healthy subjects, the salivary samples were taken to test the cortisol concentration and EEG signal recording was done simultaneously. Then, the subjects were categorized into three classes based on the salivary cortisol concentration (<5, 5-15 and >15 nmol/l). Some linear and nonlinear features extracted and finally, in order to investigate the relationship between cortisol level and EEG features, the following steps were taken on features in sequence: Genetic Algorithm, Neighboring Component Analysis, polyfit, artificial neural network and support vector machine classification. Two classifications were considered as following: state 1 categorized the subjects into three groups (three classes) and the second state put them into two groups (group 1: class 1 and 3, group 2: class 2). The best classification was done using ANN in the second state with the accuracy=94.1% while it was 92.7% in the first state. EEG features carefully predicted the cortisol level. This result is applicable to design the intelligence brain computer machines to control stress and brain performance.
Collapse
Affiliation(s)
- Behrouz Alizadeh Savareh
- National Agency for Strategic Research in Medical Education, Tehran, Iran.,Department of Health Information Management, School of Management and Medical Information Sciences, Health Human Resources Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azadeh Bashiri
- Department of Health Information Management, School of Management and Medical Information Sciences, Health Human Resources Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Hatef
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Boshra Hatef
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Islamic Republic of Iran
| |
Collapse
|
24
|
Martínez-Laorden E, Navarro-Zaragoza J, Milanés MV, Laorden ML, Almela P. Cardiac Protective Role of Heat Shock Protein 27 in the Stress Induced by Drugs of Abuse. Int J Mol Sci 2020; 21:E3623. [PMID: 32455528 PMCID: PMC7279295 DOI: 10.3390/ijms21103623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 11/16/2022] Open
Abstract
Heat shock proteins (HSP) are induced after different stress situations. Some of these proteins, particularly HSP-27, function as markers to indicate cellular stress or damage and protect the heart during addictive processes. Morphine withdrawal induces an enhancement of sympathetic activity in parallel with an increased HSP-27 expression and phosphorylation, indicating a severe situation of stress. HSP-27 can interact with different intracellular signaling pathways. Propranolol and SL-327 were able to antagonize the activation of hypothalamic-pituitary adrenal (HPA) axis and the phosphorylation of HSP-27 observed during morphine withdrawal. Therefore, β-adrenergic receptors and the extracellular signal-regulated kinase (ERK) pathway would be involved in HPA axis activity, and consequently, in HSP-27 activation. Finally, selective blockade of corticotrophin releasing factor (CRF)-1 receptor and the genetic deletion of CRF1 receptors antagonize cardiac adaptive changes. These changes are increased noradrenaline (NA) turnover, HPA axis activation and decreased HSP-27 expression and phosphorylation. This suggests a link between the HPA axis and HSP-27. On the other hand, morphine withdrawal increases µ-calpain expression, which in turn degrades cardiac troponin T (cTnT). This fact, together with a co-localization between cTnT and HSP-27, suggests that this chaperone avoids the degradation of cTnT by µ-calpain, correcting the cardiac contractility abnormalities observed during addictive processes. The aim of our research is to review the possible role of HSP-27 in the cardiac changes observed during morphine withdrawal and to understand the mechanisms implicated in its cardiac protective functions.
Collapse
Affiliation(s)
| | - Javier Navarro-Zaragoza
- Department of Pharmacology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain; (E.M.-L.); (M.V.M.); (M.L.L.); (P.A.)
| | | | | | | |
Collapse
|
25
|
Wasserman D, Wasserman J, Sokolowski M. Genetics of HPA-axis, depression and suicidality. Eur Psychiatry 2020; 25:278-80. [DOI: 10.1016/j.eurpsy.2009.12.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 12/21/2009] [Indexed: 10/19/2022] Open
Abstract
AbstractThe ultimate consequence of mental ill-health, suicidal behavior (SB), is a significant problem in most societies of the world. Suicide causes about one million deaths worldwide each year, and 10–20 times more people attempt suicide. The causes of why certain people engage in SB are complex, involving for e.g., both environmental and genetic factors, and interactions in-between. Well-established environmental risk factors are events causing significant psychological stress, which are particularly difficult to cope with, e.g. exposure to physical and sexual abuse. Excessive stress have the potential to induce unfavorable effects in a variety of higher brain-functions, incurred as side-effects to maladaptive responses in the genetically controlled stress-responsive neurosystems, e.g. the hypothalamic-pituitary-adrenal (HPA) axis; a major and systemic stress-modulator, which is mainly controlled by the regulatory corticotrophin releasing hormone receptor 1 (CRHR1) gene. Variation in-between individuals in such stress-regulatory genes such as CRHR1, may underlie the causes of the increased susceptibility of certain individuals towards SB. Here we review some of the current knowledge on what is known about the roles of the HPA axis in SB, with a focus on CRHR1.
Collapse
|
26
|
Wiersielis KR, Samuels BA, Roepke TA. Perinatal exposure to bisphenol A at the intersection of stress, anxiety, and depression. Neurotoxicol Teratol 2020; 79:106884. [PMID: 32289443 DOI: 10.1016/j.ntt.2020.106884] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022]
Abstract
Endocrine-disrupting compounds (EDCs) are common contaminants in our environment that interfere with typical endocrine function. EDCs can act on steroid and nuclear receptors or alter hormone production. One particular EDC of critical concern is bisphenol A (BPA) due to its potential harm during the perinatal period of development. Previous studies suggest that perinatal exposure to BPA alters several neurotransmitter systems and disrupts behaviors associated with depression and anxiety in the rodent offspring later in life. Thus, dysregulation in neurotransmission may translate to behavioral phenotypes observed in mood and arousal. Many of the systems disrupted by BPA also overlap with the stress system, although little evidence exists on the effects of perinatal BPA exposure in relation to stress and behavior. The purpose of this review is to explore studies involved in perinatal BPA exposure and the stress response at neurochemical and behavioral endpoints. Although more research is needed, we suggest that perinatal BPA exposure is likely inducing variations in behavioral phenotypes that modulate their action through dysregulation of neurotransmitter systems sensitive to stress and endocrine disruption.
Collapse
Affiliation(s)
- Kimberly R Wiersielis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA.
| | - Benjamin A Samuels
- Department of Psychology, School of Arts and Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ. USA
| |
Collapse
|
27
|
Thomas AW, Delevich K, Chang I, Wilbrecht L. Variation in early life maternal care predicts later long range frontal cortex synapse development in mice. Dev Cogn Neurosci 2020; 41:100737. [PMID: 31786477 PMCID: PMC6994474 DOI: 10.1016/j.dcn.2019.100737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 11/03/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022] Open
Abstract
Empirical and theoretical work suggests that early postnatal experience may inform later developing synaptic connectivity to adapt the brain to its environment. We hypothesized that early maternal experience may program the development of synaptic density on long range frontal cortex projections. To test this idea, we used maternal separation (MS) to generate environmental variability and examined how MS affected 1) maternal care and 2) synapse density on virally-labeled long range axons of offspring reared in MS or control conditions. We found that MS and variation in maternal care predicted bouton density on dorsal frontal cortex axons that terminated in the basolateral amygdala (BLA) and dorsomedial striatum (DMS) with more, fragmented care associated with higher density. The effects of maternal care on these distinct axonal projections of the frontal cortex were manifest at different ages. Maternal care measures were correlated with frontal cortex → BLA bouton density at mid-adolescence postnatal (P) day 35 and frontal cortex → DMS bouton density in adulthood (P85). Meanwhile, we found no evidence that MS or maternal care affected bouton density on ascending orbitofrontal cortex (OFC) or BLA axons that terminated in the dorsal frontal cortices. Our data show that variation in early experience can alter development in a circuit-specific and age-dependent manner that may be relevant to understanding the effects of early life adversity.
Collapse
Affiliation(s)
- A Wren Thomas
- Helen Wills Neuroscience Graduate Program, University of California, Berkeley, CA, 94720, USA
| | - Kristen Delevich
- Department of Psychology, University of California, Berkeley, CA, 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720 USA
| | - Irene Chang
- Department of Psychology, University of California, Berkeley, CA, 94720, USA
| | - Linda Wilbrecht
- Department of Psychology, University of California, Berkeley, CA, 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720 USA.
| |
Collapse
|
28
|
Yarur HE, Vega-Quiroga I, González MP, Noches V, Thomases DR, Andrés ME, Ciruela F, Tseng KY, Gysling K. Inhibitory Control of Basolateral Amygdalar Transmission to the Prefrontal Cortex by Local Corticotrophin Type 2 Receptor. Int J Neuropsychopharmacol 2019; 23:108-116. [PMID: 31800046 PMCID: PMC7094000 DOI: 10.1093/ijnp/pyz065] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/01/2019] [Accepted: 12/03/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Basolateral amygdalar projections to the prefrontal cortex play a key role in modulating behavioral responses to stress stimuli. Among the different neuromodulators known to impact basolateral amygdalar-prefrontal cortex transmission, the corticotrophin releasing factor (CRF) is of particular interest because of its role in modulating anxiety and stress-associated behaviors. While CRF type 1 receptor (CRFR1) has been involved in prefrontal cortex functioning, the participation of CRF type 2 receptor (CRFR2) in basolateral amygdalar-prefrontal cortex synaptic transmission remains unclear. METHODS Immunofluorescence anatomical studies using rat prefrontal cortex synaptosomes devoid of postsynaptic elements were performed in rats with intra basolateral amygdalar injection of biotinylated dextran amine. In vivo microdialysis and local field potential recordings were used to measure glutamate extracellular levels and changes in long-term potentiation in prefrontal cortex induced by basolateral amygdalar stimulation in the absence or presence of CRF receptor antagonists. RESULTS We found evidence for the presynaptic expression of CRFR2 protein and mRNA in prefrontal cortex synaptic terminals originated from basolateral amygdalar. By means of microdialysis and electrophysiological recordings in combination with an intra-prefrontal cortex infusion of the CRFR2 antagonist antisauvagine-30, we were able to determine that CRFR2 is functionally positioned to limit the strength of basolateral amygdalar transmission to the prefrontal cortex through presynaptic inhibition of glutamate release. CONCLUSIONS Our study shows for the first time to our knowledge that CRFR2 is expressed in basolateral amygdalar afferents projecting to the prefrontal cortex and exerts an inhibitory control of prefrontal cortex responses to basolateral amygdalar inputs. Thus, changes in CRFR2 signaling are likely to disrupt the functional connectivity of the basolateral amygdalar-prefrontal cortex pathway and associated behavioral responses.
Collapse
Affiliation(s)
- Hector E Yarur
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Vega-Quiroga
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcela P González
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Verónica Noches
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel R Thomases
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - María E Andrés
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois,Correspondence: Katia Gysling, PhD, Department of Cellular and Molecular Biology Faculty of Biological Sciences Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile (); Kuei Y. Tseng, PhD, Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA ()
| | - Katia Gysling
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile,Correspondence: Katia Gysling, PhD, Department of Cellular and Molecular Biology Faculty of Biological Sciences Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile (); Kuei Y. Tseng, PhD, Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA ()
| |
Collapse
|
29
|
Pandey GN, Rizavi HS, Bhaumik R, Ren X. Increased protein and mRNA expression of corticotropin-releasing factor (CRF), decreased CRF receptors and CRF binding protein in specific postmortem brain areas of teenage suicide subjects. Psychoneuroendocrinology 2019; 106:233-243. [PMID: 31005044 PMCID: PMC7061258 DOI: 10.1016/j.psyneuen.2019.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/07/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022]
Abstract
Overactivity of hypothalamic-pituitary-adrenal (HPA) axis function has been implicated in depression and suicidal behavior. This is based on the observation of an abnormal dexamethasone (DEX) and DEX-adrenocorticotropic hormone (ACTH) test in patients with depression and suicidal behavior. Recently, some studies have also found abnormalities of glucocorticoid receptors (GR), mineralocorticoid receptors (MR), corticotropin releasing factor (CRF), CRF receptors (CRF-R) and CRF binding protein (CRF-BP) in depressed and suicidal patients. Some investigators have also observed increased levels of CRF in the cerebrospinal fluid (CSF) and altered levels of MR, GR and CRF in the postmortem brain of depressed and suicidal subjects. We have earlier reported decreased protein and mRNA expression of GR and GILZ, a chaperone protein, in the postmortem brain of teenage suicide subjects. We have further studied CRF and its receptors in different areas of the postmortem brain of suicide subjects, i.e., the prefrontal cortex (PFC), hippocampus (HIPPO), subiculum and amygdala (AMY) from teenage suicide subjects. The CRF and its receptors were determined in the PFC (Brodmann area 9), HIPPO, subiculum and different amygdaloid nuclei from 24 normal control subjects and 24 teenage suicide subjects. Protein expression of CRF, its receptors and CRF-BP was determined by immunolabeling using the Western blot technique and mRNA expression was determined by real-time PCR (qPCR) technique. We found that the mRNA levels of CRF were significantly increased in the PFC, in the central amygdaloid nucleus (CeAMY) and in the subiculum. mRNA levels of CRF-R1 and CRF-BP were significantly decreased in the PFC. We did not find any changes in the HIPPO of any of the CRF components we studied. When we compared the protein expression of CRF components we found that CRF was significantly increased and CRF-R1, CRF-R2 and CRF-BP significantly decreased in the PFC. On the other hand, there were no changes in the protein expression of CRF components in the HIPPO. Our results in the postmortem brain suggest that, as found by clinical studies in the CSF, there are significant alterations of CRF and its receptors in the postmortem brain of teenage suicide subjects. These alterations of CRF and its components were region-specific, as changes were not generally observed in the HIPPO.
Collapse
Affiliation(s)
- Ghanshyam N. Pandey
- Corresponding Author: Ghanshyam N. Pandey, Ph.D., University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612, USA, Phone (312) 413-4540, Fax: (312) 413-4547,
| | | | | | | |
Collapse
|
30
|
Fischl MJ, Ueberfuhr MA, Drexl M, Pagella S, Sinclair JL, Alexandrova O, Deussing JM, Kopp-Scheinpflug C. Urocortin 3 signalling in the auditory brainstem aids recovery of hearing after reversible noise-induced threshold shift. J Physiol 2019; 597:4341-4355. [PMID: 31270820 PMCID: PMC6852351 DOI: 10.1113/jp278132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/03/2019] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Ongoing, moderate noise exposure does not instantly damage the auditory system but may cause lasting deficits, such as elevated thresholds and accelerated ageing of the auditory system. The neuromodulatory peptide urocortin-3 (UCN3) is involved in the body's recovery from a stress response, and is also expressed in the cochlea and the auditory brainstem. Lack of UCN3 facilitates age-induced hearing loss and causes permanently elevated auditory thresholds following a single 2 h noise exposure at moderate intensities. Outer hair cell function in mice lacking UCN3 is unaffected, so that the observed auditory deficits are most likely due to inner hair cell function or central mechanisms. Highly specific, rather than ubiquitous, expression of UCN3 in the brain renders it a promising candidate for designing drugs to ameliorate stress-related auditory deficits, including recovery from acoustic trauma. ABSTRACT Environmental acoustic noise is omnipresent in our modern society, with sound levels that are considered non-damaging still causing long-lasting or permanent changes in the auditory system. The small neuromodulatory peptide urocortin-3 (UCN3) is the endogenous ligand for corticotropin-releasing factor receptor type 2 and together they are known to play an important role in stress recovery. UCN3 expression has been observed in the auditory brainstem, but its role remains unclear. Here we describe the detailed distribution of UCN3 expression in the murine auditory brainstem and provide evidence that UCN3 is expressed in the synaptic region of inner hair cells in the cochlea. We also show that mice with deficient UCN3 signalling experience premature ageing of the auditory system starting at an age of 4.7 months with significantly elevated thresholds of auditory brainstem responses (ABRs) compared to age-matched wild-type mice. Following a single, 2 h exposure to moderate (84 or 94 dB SPL) noise, UCN3-deficient mice exhibited significantly larger shifts in ABR thresholds combined with maladaptive recovery. In wild-type mice, the same noise exposure did not cause lasting changes to auditory thresholds. The presence of UCN3-expressing neurons throughout the auditory brainstem and the predisposition to hearing loss caused by preventing its normal expression suggests UCN3 as an important neuromodulatory peptide in the auditory system's response to loud sounds.
Collapse
Affiliation(s)
- Matthew J Fischl
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| | - Margarete A Ueberfuhr
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Munich, Germany
| | - Markus Drexl
- German Center for Vertigo and Balance Disorders, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Sara Pagella
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Munich, Germany
| | - James L Sinclair
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| | - Olga Alexandrova
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| | - Jan M Deussing
- Max Planck Institute of Psychiatry, Molecular Neurogenetics, Munich, Germany
| | - Conny Kopp-Scheinpflug
- Department of Biology II, Division Neurobiology, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
31
|
Navarro G, Medrano M, Aguinaga D, Vega-Quiroga I, Lillo A, Jiménez J, Casanovas M, Canela EI, Mallol J, Gysling K, Franco R. Differential effect of amphetamine over the corticotropin-releasing factor CRF 2 receptor, the orexin OX 1 receptor and the CRF 2-OX 1 heteroreceptor complex. Neuropharmacology 2018; 152:102-111. [PMID: 30465812 DOI: 10.1016/j.neuropharm.2018.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/16/2018] [Accepted: 11/09/2018] [Indexed: 11/30/2022]
Abstract
Stress is one of the factors underlying drug seeking behavior that often goes in parallel with loss of appetite. We here demonstrate that orexin 1 receptors (OX1R) may form complexes with the corticotropin releasing factor CRF2 receptor. Two specific features of the heteromer were a cross-antagonism and a blockade by CRF2 of OX1R signaling. In cells expressing one of the receptors, agonist-mediated signal transduction mechanisms were potentiated by amphetamine. Sigma 1 (σ1) and 2 (σ2) receptors are targets of drugs of abuse and, despite sharing a similar name, the two receptors are structurally unrelated and their physiological role is not known. We here show that σ1 receptors interact with CRF2 receptors and that σ2 receptors interact with OX1R. Moreover, we show that amphetamine effect on CRF2 receptors was mediated by σ1R whereas the effect on OX1 receptors was mediated by σ2R. Amphetamine did potentiate the negative cross-talk occurring within the CRF2-OX1 receptor heteromer context, likely by a macromolecular complex involving the two sigma receptors and the two GPCRs. Finally, in vivo microdialysis experiments showed that amphetamine potentiated orexin A-induced dopamine and glutamate release in the ventral tegmental area (VTA). Remarkably, the in vivo orexin A effects were blocked by a selective CRF2R antagonist. These results show that amphetamine impacts on the OX1R-, CRF2R- and OX1R/CRF2R-mediated signaling and that cross-antagonism is instrumental for in vivo detection of GPCR heteromers. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Gemma Navarro
- Department of Biochemistry and Physiology, Pharmacy and Food Science School, University of Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mireia Medrano
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| | - David Aguinaga
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| | - Ignacio Vega-Quiroga
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro Lillo
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| | - Jasmina Jiménez
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mireia Casanovas
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| | - Enric I Canela
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| | - Josefa Mallol
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| | - Katia Gysling
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rafael Franco
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain.
| |
Collapse
|
32
|
Lam VY, Raineki C, Ellis L, Yu W, Weinberg J. Interactive effects of prenatal alcohol exposure and chronic stress in adulthood on anxiety-like behavior and central stress-related receptor mRNA expression: Sex- and time-dependent effects. Psychoneuroendocrinology 2018; 97:8-19. [PMID: 29990678 PMCID: PMC6424330 DOI: 10.1016/j.psyneuen.2018.06.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/18/2018] [Accepted: 06/22/2018] [Indexed: 01/24/2023]
Abstract
Children and adults prenatally exposed to alcohol show higher rates of mental health problems than unexposed individuals, with depression and anxiety being among the more commonly encountered disorders. Previous studies in rats showed that prenatal alcohol exposure (PAE) can indeed increase depressive- and anxiety-like behavior in adulthood; however, depression and anxiety are often observed in the context of stress and/or a dysregulated stress response system (the hypothalamic-pituitary-adrenal [HPA] axis). PAE can dysregulate the HPA axis, resulting in hyperresponsivity to stress. In turn, this may predispose individuals prenatally exposed to alcohol to the adverse effects of stress compared to unexposed individuals. We have shown previously that PAE animals may be more sensitive to the effects of chronic stress on behavior, showing increased anxiety- and depressive-like behavior following chronic unpredictable stress (CUS) exposure. Here, we investigated the independent and interactive effects of PAE and adult CUS on anxiety-like behavior and receptor systems (corticotropin-releasing hormone receptor type 1 [CRHR1], mineralocorticoid receptor [MR], and glucocorticoid receptor [GR]), and underlying stress and emotional regulation, and whether exposure to CUS differentially results in immediate or delayed effects. Adult male and female offspring from PAE, pair-fed (PF), and ad libitum-fed control (C) dams were exposed to either 10 days of CUS or left undisturbed. Behavioral testing began 1 or 14 days post-CUS, and brains were collected following testing. Anxiety-like behaviors were evaluated using the open field, elevated plus maze and dark-light emergence tests. CRHR1, MR, and GR mRNA expression were assessed in the medial prefrontal cortex (mPFC), amygdala, and hippocampal formation, brain areas key to both stress and emotional regulation. We found that PAE differentially increased anxiety-like behavior and altered GR mRNA in males and females compared to their control counterparts. Furthermore, depending on the timing of testing, CUS unmasked alterations in GR and CRHR1 mRNA expression in the mPFC and amygdala in PAE males, and MR mRNA in the hippocampal formation in PAE females compared to their C counterparts. Overall, the changes observed in these receptor systems may underlie the increase in anxiety-like behavior following PAE and CUS exposure in adulthood. That CUS differentially affected brain and behavioral outcome of PAE and C animals, and did so in a sexually-dimorphic manner, has important implications for understanding the etiology of psychopathology in individuals prenatally exposed to alcohol.
Collapse
Affiliation(s)
- Vivian Y.Y. Lam
- Corresponding author at: Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada. (V.Y.Y. Lam)
| | | | | | | | | |
Collapse
|
33
|
Deussing JM, Chen A. The Corticotropin-Releasing Factor Family: Physiology of the Stress Response. Physiol Rev 2018; 98:2225-2286. [DOI: 10.1152/physrev.00042.2017] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The physiological stress response is responsible for the maintenance of homeostasis in the presence of real or perceived challenges. In this function, the brain activates adaptive responses that involve numerous neural circuits and effector molecules to adapt to the current and future demands. A maladaptive stress response has been linked to the etiology of a variety of disorders, such as anxiety and mood disorders, eating disorders, and the metabolic syndrome. The neuropeptide corticotropin-releasing factor (CRF) and its relatives, the urocortins 1–3, in concert with their receptors (CRFR1, CRFR2), have emerged as central components of the physiological stress response. This central peptidergic system impinges on a broad spectrum of physiological processes that are the basis for successful adaptation and concomitantly integrate autonomic, neuroendocrine, and behavioral stress responses. This review focuses on the physiology of CRF-related peptides and their cognate receptors with the aim of providing a comprehensive up-to-date overview of the field. We describe the major molecular features covering aspects of gene expression and regulation, structural properties, and molecular interactions, as well as mechanisms of signal transduction and their surveillance. In addition, we discuss the large body of published experimental studies focusing on state-of-the-art genetic approaches with high temporal and spatial precision, which collectively aimed to dissect the contribution of CRF-related ligands and receptors to different levels of the stress response. We discuss the controversies in the field and unravel knowledge gaps that might pave the way for future research directions and open up novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jan M. Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
34
|
Kreouzis V, Chen GL, Miller GM. Perturbations of Neuron-Restrictive Silencing Factor Modulate Corticotropin-Releasing Hormone Gene Expression in the Human Cell Line BeWo. MOLECULAR NEUROPSYCHIATRY 2018; 4:100-110. [PMID: 30397598 DOI: 10.1159/000492635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/02/2018] [Indexed: 01/04/2023]
Abstract
Stress exacerbates disease, and understanding its molecular mechanisms is crucial to the development of novel therapeutic interventions to combat stress-related disorders. The driver of the stress response in the hypothalamic-pituitary-adrenal axis (HPA) is corticotropin-releasing hormone (CRH), a neuropeptide synthesized in the paraventricular nucleus of the hypothalamus. Evidence supports that CRH expression is epigenetically modified at the molecular level by environmental stimuli, causing changes in the stress response. This effect is mediated by a concert of factors that translate environmental change into alterations in gene expression. An important regulator and epigenetic modulator of CRH expression is neuron-restrictive silencing factor (NRSF). Previously, our lab identified numerous splice variants of NRSF that are specific to humans and predictive of differential regulatory effects of NRSF variants on targeted gene expression. The human cell line BeWo has endogenous CRH and NRSF expression providing an in vitro model system. Here, we show that manipulation of NRSF expression through siRNA technology, overexpression by plasmid vectors, and direct cAMP induction that CRH expression is linked to changes in NRSF expression. Accordingly, this epigenetic regulatory pathway in humans might be a critical mechanism involved in the regulation of the stress response.
Collapse
Affiliation(s)
- Vasileios Kreouzis
- Department of Psychology, College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Guo-Lin Chen
- Department of Pharmaceutical Sciences, Bouve College of Health Sciences, Boston, Massachusetts, USA
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Bouve College of Health Sciences, Boston, Massachusetts, USA.,Department of Chemical Engineering, College of Engineering, Boston, Massachusetts, USA.,Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Fujii Y, Suzuki K, Hasegawa Y, Nanba F, Toda T, Adachi T, Taira S, Osakabe N. Single oral administration of flavan 3-ols induces stress responses monitored with stress hormone elevations in the plasma and paraventricular nucleus. Neurosci Lett 2018; 682:106-111. [PMID: 29902479 DOI: 10.1016/j.neulet.2018.06.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/16/2018] [Accepted: 06/08/2018] [Indexed: 02/05/2023]
Abstract
We previously confirmed that postprandial alterations in the circulation and metabolism after a single oral dose of flavan 3-ols (mixture of catechin and catechin oligomers) were involved in an increase in sympathetic nervous activity. However, it is well known that, in response to various stresses, activation of the hypothalamic-pituitary-adrenal (HPA) axis occurs together with sympathetic nerve activity, which is associated with activation of the sympathetic-adrenal-medullary (SAM) axis. In this study, we examined whether the HPA axis was activated after a single dose of flavan 3-ols. We administered an oral dose of 10 or 50 mg/kg flavan 3-ols to male ICR mice, removed the brains, and fixed them in paraformaldehyde-phosphate buffer. Other animals that were treated similarly were decapitated, and blood was collected. In the paraventricular nucleus (PVN), c-fos mRNA expression increased significantly at 15 min after administration of either 10 or 50 mg/kg flavan 3-ols. Corticotropin-releasing hormone (CRH) mRNA expression levels significantly increased at 240 min after administration of 10 mg/kg flavan 3-ols, and at 60 min after administration of 50 mg/kg flavan 3-ols. Plasma corticosterone levels were also significantly increased at 240 min after ingestion of 50 mg/kg flavan 3-ols. In this experiment, we confirmed that the ingestion of flavan 3-ols acted as a stressor in mammals with activation both the SAM and HPA axes.
Collapse
Affiliation(s)
- Yasuyuki Fujii
- Department of Bioscience and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Munumaku, Saitama, 337-8570, Japan
| | - Kenta Suzuki
- Department of Bioscience and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Munumaku, Saitama, 337-8570, Japan
| | - Yahiro Hasegawa
- Department of Bioscience and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Munumaku, Saitama, 337-8570, Japan
| | - Fumio Nanba
- Department of Research and Development, Fujicco Co. Ltd. Hyogo, 650-8558, Japan
| | - Toshiya Toda
- Department of Research and Development, Fujicco Co. Ltd. Hyogo, 650-8558, Japan
| | - Takahiro Adachi
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, 113-8510, Japan
| | - Shu Taira
- Fukushima University, Faculty of Food and Agricultural Sciences, Kanayagawa, Fukushima, 960-1248, Japan
| | - Naomi Osakabe
- Department of Bioscience and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Munumaku, Saitama, 337-8570, Japan.
| |
Collapse
|
36
|
Kelly EA, Fudge JL. The neuroanatomic complexity of the CRF and DA systems and their interface: What we still don't know. Neurosci Biobehav Rev 2018; 90:247-259. [PMID: 29704516 PMCID: PMC5993645 DOI: 10.1016/j.neubiorev.2018.04.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/14/2018] [Accepted: 04/15/2018] [Indexed: 12/28/2022]
Abstract
Corticotropin-releasing factor (CRF) is a neuropeptide that mediates the stress response. Long known to contribute to regulation of the adrenal stress response initiated in the hypothalamic-pituitary axis (HPA), a complex pattern of extrahypothalamic CRF expression is also described in rodents and primates. Cross-talk between the CRF and midbrain dopamine (DA) systems links the stress response to DA regulation. Classically CRF + cells in the extended amygdala and paraventricular nucleus (PVN) are considered the main source of this input, principally targeting the ventral tegmental area (VTA). However, the anatomic complexity of both the DA and CRF system has been increasingly elaborated in the last decade. The DA neurons are now recognized as having diverse molecular, connectional and physiologic properties, predicted by their anatomic location. At the same time, the broad distribution of CRF cells in the brain has been increasingly delineated using different species and techniques. Here, we review updated information on both CRF localization and newer conceptualizations of the DA system to reconsider the CRF-DA interface.
Collapse
Affiliation(s)
- E A Kelly
- University of Rochester, School of Medicine and Dentistry, The Ernest J Del Monte Institute for Neuroscience, Department of Neuroscience, Rochester, NY, United States
| | - J L Fudge
- University of Rochester, School of Medicine and Dentistry, The Ernest J Del Monte Institute for Neuroscience, Department of Neuroscience, Rochester, NY, United States; University of Rochester, School of Medicine and Dentistry, The Ernest J Del Monte Institute for Neuroscience, Department of Psychiatry, Rochester, NY, United States.
| |
Collapse
|
37
|
Yarushkina NI, Filaretova LP. Corticotropin-Releasing Factor (CRF) and Somatic Pain Sensitivity: the Contribution of CRF Receptors of Subtypes 1 and 2. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Leistner C, Menke A. How to measure glucocorticoid receptor's sensitivity in patients with stress-related psychiatric disorders. Psychoneuroendocrinology 2018; 91:235-260. [PMID: 29449045 DOI: 10.1016/j.psyneuen.2018.01.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 12/07/2017] [Accepted: 01/28/2018] [Indexed: 12/31/2022]
Abstract
Stress is a state of derailed homeostasis and a main environmental risk factor for psychiatric diseases. Chronic or uncontrollable stress may lead to a dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, which is a common feature of stress-related psychiatric disorders. One of the key mechanisms underlying a disturbed HPA axis is an impaired function of the glucocorticoid receptor (GR) with an enhanced or reduced feedback sensitivity for glucocorticoids and subsequently altered concentrations of peripheral cortisol. GR function is regulated by a multiprotein complex including the different expression of the hsp90 co-chaperone FK 506 binding protein 51 (FKBP5) that may be genetically determined or acquired in response to stressful stimuli. Specific patterns of a dysregulation of the HPA axis and GR function are found in different stress-related psychiatric entities e.g. major depression, job-related exhaustion or posttraumatic stress disorder. GR challenge tests like the dexamethasone-suppression test (DST), the dexamethasone-corticotropin-releasing hormone (dex-CRH) test or most recently the analysis of the dexamethasone-induced gene expression are employed to sensitively measure HPA axis activity in these disorders. They provide information for a stratification of phenotypic similar but neurobiological diverse psychiatric disorders. In this review we present a synopsis of GR challenge tests with a focus on the application of the DST, the CRH test and the dex-CRH test as well as the dexamethasone-induced gene expression in stress-related psychiatric entities.
Collapse
Affiliation(s)
- Carolin Leistner
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Margarete-Hoeppel-Platz 1, Wuerzburg, 97080, Germany
| | - Andreas Menke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Margarete-Hoeppel-Platz 1, Wuerzburg, 97080, Germany; Comprehensive Heart Failure Center, University Hospital of Wuerzburg, Am Schwarzenberg 15, Wuerzburg, 97080, Germany.
| |
Collapse
|
39
|
Dong H, Keegan JM, Hong E, Gallardo C, Montalvo-Ortiz J, Wang B, Rice KC, Csernansky J. Corticotrophin releasing factor receptor 1 antagonists prevent chronic stress-induced behavioral changes and synapse loss in aged rats. Psychoneuroendocrinology 2018; 90:92-101. [PMID: 29477954 PMCID: PMC5864558 DOI: 10.1016/j.psyneuen.2018.02.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/25/2018] [Accepted: 02/14/2018] [Indexed: 11/24/2022]
Abstract
Mounting evidence suggests that chronic stress can alter brain structure and function and promote the development of neuropsychiatric disorders, such as depression and Alzheimer's disease. Although the results of several studies have indicated that aged brains are more vulnerable to chronic stress, it remains unknown whether antagonists of a key stress regulator, the corticotrophin releasing factor receptor 1 (CRF1), can prevent stress-induced anxiety and memory deficits in animal models. In this study, we evaluated the potential benefits of two CRF1 antagonists, R121919 and antalarmin, for preventing stress-induced anxiety-related behavioral and memory deficits and neurodegeneration in aged rats. We stressed rats using isolation-restraint for 3 months starting from the 18 months of age. Subsets of animals were administrated either R121919 or antalarmin through food chow for 3 months, followed by a series of behavioral, biochemical and morphological analyses. We found that stressed aged rats displayed body weight losses and increased corticosterone levels, as well as anxiety-related behaviors and memory deficits. Additionally, chronic stress induced a loss of cortical dendritic spines and synapses. However, R121919 and antalarmin both prevented stress-induced behavioral changes including anxiety-related behaviors and memory deficits and prevented synapse loss, perhaps through reversing HPA axis dysfunction. These results suggest that CRF1 antagonists may hold promise as a potential therapy for preventing stress-induced anxiety and memory deficits in aged individuals.
Collapse
Affiliation(s)
- Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| | - Jack M Keegan
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Ellie Hong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Christopher Gallardo
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Janitza Montalvo-Ortiz
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Becky Wang
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Kenner C. Rice
- National Institute on Drug Abuse, and National Institute Alcohol Abuse and Alcoholism Intramural Research Program, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - John Csernansky
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|
40
|
Yan Y, Dominguez S, Fisher DW, Dong H. Sex differences in chronic stress responses and Alzheimer's disease. Neurobiol Stress 2018; 8:120-126. [PMID: 29888307 PMCID: PMC5991323 DOI: 10.1016/j.ynstr.2018.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/10/2018] [Accepted: 03/17/2018] [Indexed: 11/13/2022] Open
Abstract
Clinical studies indicate that Alzheimer's disease (AD) disproportionately affects women in both disease prevalence and severity, but the mechanisms underlying this sex divergence are unknown. Though some have suggested this difference in risk is a reflection of known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities towards the disease. While a number of potential risk factors have been hypothesized to affect these differences in risks, none have been definitively verified. In this review, we discuss a novel hypothesis whereby women's susceptibility to chronic stress also mediates increased risk for AD. As stress is a risk factor for AD, and women are twice as likely to develop mood disorders where stress is a major etiology, it is possible that sex dimorphisms in stress responses contribute to the increase in women with AD. In line with this, sex divergence in biochemical responses to stress have been noted along the hypothalamic-pituitary-adrenal (HPA) axis and among known molecular effectors of AD, with crosstalk between these processes also being likely. In addition, activation of the cortical corticotrophin-releasing factor receptor 1 (CRF1) signaling pathway leads to distinct female-biased increases in molecules associated with AD pathogenesis. Therefore, the different biochemical responses to stress between women and men may represent an intrinsic, sex-dependent risk factor for AD.
Collapse
Affiliation(s)
- Yan Yan
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Department of Physiology, Zunyi Medical University, Zunyi Guizhou 563099, China
| | - Sky Dominguez
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Daniel W. Fisher
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Department of Physiology, Zunyi Medical University, Zunyi Guizhou 563099, China
| |
Collapse
|
41
|
Henckens MJAG, Printz Y, Shamgar U, Dine J, Lebow M, Drori Y, Kuehne C, Kolarz A, Eder M, Deussing JM, Justice NJ, Yizhar O, Chen A. CRF receptor type 2 neurons in the posterior bed nucleus of the stria terminalis critically contribute to stress recovery. Mol Psychiatry 2017; 22:1691-1700. [PMID: 27550842 DOI: 10.1038/mp.2016.133] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 05/24/2016] [Accepted: 06/01/2016] [Indexed: 12/20/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is critical in mediating states of anxiety, and its dysfunction has been linked to stress-related mental disease. Although the anxiety-related role of distinct subregions of the anterior BNST was recently reported, little is known about the contribution of the posterior BNST (pBNST) to the behavioral and neuroendocrine responses to stress. Previously, we observed abnormal expression of corticotropin-releasing factor receptor type 2 (CRFR2) to be associated with post-traumatic stress disorder (PTSD)-like symptoms. Here, we found that CRFR2-expressing neurons within the pBNST send dense inhibitory projections to other stress-related brain regions (for example, the locus coeruleus, medial amygdala and paraventricular nucleus), implicating a prominent role of these neurons in orchestrating the neuroendocrine, autonomic and behavioral response to stressful situations. Local CRFR2 activation by urocortin 3 depolarized the cells, increased the neuronal input resistance and increased firing of action potentials, indicating an enhanced excitability. Furthermore, we showed that CRFR2-expressing neurons within the pBNST are critically involved in the modulation of the behavioral and neuroendocrine response to stress. Optogenetic activation of CRFR2 neurons in the pBNST decreased anxiety, attenuated the neuroendocrine stress response, ameliorated stress-induced anxiety and impaired the fear memory for the stressful event. Moreover, activation following trauma exposure reduced the susceptibility for PTSD-like symptoms. Optogenetic inhibition of pBNST CRFR2 neurons yielded opposite effects. These data indicate the relevance of pBNST activity for adaptive stress recovery.
Collapse
Affiliation(s)
- M J A G Henckens
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Y Printz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - U Shamgar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - J Dine
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - M Lebow
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Y Drori
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - C Kuehne
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A Kolarz
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - M Eder
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - J M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - N J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - O Yizhar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - A Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
42
|
Greenwood-Van Meerveld B, Johnson AC. Stress-Induced Chronic Visceral Pain of Gastrointestinal Origin. Front Syst Neurosci 2017; 11:86. [PMID: 29213232 PMCID: PMC5702626 DOI: 10.3389/fnsys.2017.00086] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
Visceral pain is generally poorly localized and characterized by hypersensitivity to a stimulus such as organ distension. In concert with chronic visceral pain, there is a high comorbidity with stress-related psychiatric disorders including anxiety and depression. The mechanisms linking visceral pain with these overlapping comorbidities remain to be elucidated. Evidence suggests that long term stress facilitates pain perception and sensitizes pain pathways, leading to a feed-forward cycle promoting chronic visceral pain disorders such as irritable bowel syndrome (IBS). Early life stress (ELS) is a risk-factor for the development of IBS, however the mechanisms responsible for the persistent effects of ELS on visceral perception in adulthood remain incompletely understood. In rodent models, stress in adult animals induced by restraint and water avoidance has been employed to investigate the mechanisms of stress-induce pain. ELS models such as maternal separation, limited nesting, or odor-shock conditioning, which attempt to model early childhood experiences such as neglect, poverty, or an abusive caregiver, can produce chronic, sexually dimorphic increases in visceral sensitivity in adulthood. Chronic visceral pain is a classic example of gene × environment interaction which results from maladaptive changes in neuronal circuitry leading to neuroplasticity and aberrant neuronal activity-induced signaling. One potential mechanism underlying the persistent effects of stress on visceral sensitivity could be epigenetic modulation of gene expression. While there are relatively few studies examining epigenetically mediated mechanisms involved in visceral nociception, stress-induced visceral pain has been linked to alterations in DNA methylation and histone acetylation patterns within the brain, leading to increased expression of pro-nociceptive neurotransmitters. This review will discuss the potential neuronal pathways and mechanisms responsible for stress-induced exacerbation of chronic visceral pain. Additionally, we will review the importance of specific experimental models of adult stress and ELS in enhancing our understanding of the basic molecular mechanisms of pain processing.
Collapse
Affiliation(s)
- Beverley Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
- VA Medical Center, Oklahoma City, OK, United States
| | | |
Collapse
|
43
|
Bishop SA, Dech RT, Guzik P, Neary JP. Heart rate variability and implication for sport concussion. Clin Physiol Funct Imaging 2017; 38:733-742. [PMID: 29144026 DOI: 10.1111/cpf.12487] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 10/19/2017] [Indexed: 12/12/2022]
Abstract
Finding sensitive and specific markers for sports-related concussion is both challenging and clinically important. Such biomarkers might be helpful in the management of patients with concussion (i.e. diagnosis, monitoring and risk prediction). Among many parameters, blood flow-pressure metrics and heart rate variability (HRV) have been used to gauge concussion outcomes. Reports on the relation between HRV and both acute and prolonged concussion recovery are conflicting. While some authors report on differences in the low-frequency (LF) component of HRV during postural manipulations and postexercise conditions, others observe no significant differences in various HRV measures. Despite the early success of using the HRV LF for concussion recovery, the interpretation of the LF is debated. Recent research suggests the LF power is a net effect of several intrinsic modulatory factors from both sympathetic and parasympathetic branches of the autonomic nervous system, vagally mediated baroreflex and even some respiratory influences at lower respiratory rate. There are only a few well-controlled concussion studies that specifically examine the contribution of the autonomic nervous system branches with HRV for concussion management. This study reviews the most recent HRV- concussion literature and the underlying HRV physiology. It also highlights cerebral blood flow studies related to concussion and the importance of multimodal assessment of various biological signals. It is hoped that a better understanding of the physiology behind HRV might generate cost-effective, repeatable and reliable protocols, all of which will improve the interpretation of HRV throughout concussion recovery.
Collapse
Affiliation(s)
- Scott A Bishop
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, Saskatchewan, Canada
| | - Ryan T Dech
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, Saskatchewan, Canada
| | - Przemyslaw Guzik
- Department of Cardiology-Intensive Care Therapy & Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - J Patrick Neary
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, Saskatchewan, Canada
| |
Collapse
|
44
|
Varodayan FP, Correia D, Kirson D, Khom S, Oleata CS, Luu G, Schweitzer P, Roberto M. CRF modulates glutamate transmission in the central amygdala of naïve and ethanol-dependent rats. Neuropharmacology 2017; 125:418-428. [PMID: 28807676 DOI: 10.1016/j.neuropharm.2017.08.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/21/2017] [Accepted: 08/09/2017] [Indexed: 11/15/2022]
Abstract
Corticotropin-releasing factor (CRF) signaling in the central nucleus of the amygdala (CeA) is hypothesized to drive the development of alcohol dependence, as it regulates ethanol intake and several anxiogenic behaviors linked to withdrawal. Excitatory glutamatergic neurotransmission contributes to alcohol reinforcement, tolerance and dependence. Therefore, in this study we used in vitro slice electrophysiology to investigate the effects of CRF and its receptor subtype (CRF1 and CRF2) antagonists on both evoked and spontaneous action potential-independent glutamatergic transmission in the CeA of naive and ethanol-dependent Sprague-Dawley rats. We found that CRF (25-200 nM) concentration-dependently diminished evoked compound excitatory postsynaptic potentials (EPSPs), but increased miniature excitatory postsynaptic current (mEPSC) frequencies similarly in CeA neurons of both naïve and ethanol-dependent rats, indicating reduced evoked glutamatergic responses and enhanced vesicular glutamate release, respectively. This CRF-induced vesicular glutamate release was prevented by the CRF1/2 antagonist (Astressin B) and the CRF1 antagonist (R121919), but not by the CRF2 antagonist (Astressin 2B). Similarly, CRF's effects on evoked glutamatergic responses were completely blocked by CRF1 antagonism, but only slightly decreased in the presence of the CRF2 antagonist. Moreover, CRF1 antagonism reveals a tonic facilitation of vesicular glutamate, whereas the CRF2 antagonism revealed a tonic inhibition of vesicular glutamate release. Collectively our data show that CRF primarily acts at presynaptic CRF1 to produce opposite effects on CeA evoked and spontaneous glutamate release and that the CRF system modulates CeA glutamatergic synapses throughout the development of alcohol dependence.
Collapse
Affiliation(s)
| | - Diego Correia
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA; Department of Pharmacology, Universidade Federal do Paraná, Jardim das Américas, Curitiba, Paraná, CEP 81531-990, Brazil
| | - Dean Kirson
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Sophia Khom
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | | | - George Luu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Paul Schweitzer
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
45
|
Roberto M, Spierling SR, Kirson D, Zorrilla EP. Corticotropin-Releasing Factor (CRF) and Addictive Behaviors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:5-51. [PMID: 29056155 PMCID: PMC6155477 DOI: 10.1016/bs.irn.2017.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Drug addiction is a complex disorder that is characterized by compulsivity to seek and take the drug, loss of control in limiting intake of the drug, and emergence of a withdrawal syndrome in the absence of the drug. The transition from casual drug use to dependence is mediated by changes in reward and brain stress functions and has been linked to a shift from positive reinforcement to negative reinforcement. The recruitment of brain stress systems mediates the negative emotional state produced by dependence that drives drug seeking through negative reinforcement mechanisms, defined as the "dark side" of addiction. In this chapter we focus on behavioral and cellular neuropharmacological studies that have implicated brain stress systems (i.e., corticotropin-releasing factor [CRF]) in the transition to addiction and the predominant brain regions involved. We also discuss the implication of CRF recruitment in compulsive eating disorders.
Collapse
Affiliation(s)
- Marisa Roberto
- The Scripps Research Institute, La Jolla, CA, United States.
| | | | - Dean Kirson
- The Scripps Research Institute, La Jolla, CA, United States
| | | |
Collapse
|
46
|
Inda C, Armando NG, Dos Santos Claro PA, Silberstein S. Endocrinology and the brain: corticotropin-releasing hormone signaling. Endocr Connect 2017; 6:R99-R120. [PMID: 28710078 PMCID: PMC5551434 DOI: 10.1530/ec-17-0111] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 01/01/2023]
Abstract
Corticotropin-releasing hormone (CRH) is a key player of basal and stress-activated responses in the hypothalamic-pituitary-adrenal axis (HPA) and in extrahypothalamic circuits, where it functions as a neuromodulator to orchestrate humoral and behavioral adaptive responses to stress. This review describes molecular components and cellular mechanisms involved in CRH signaling downstream of its G protein-coupled receptors (GPCRs) CRHR1 and CRHR2 and summarizes recent findings that challenge the classical view of GPCR signaling and impact on our understanding of CRHRs function. Special emphasis is placed on recent studies of CRH signaling that revealed new mechanistic aspects of cAMP generation and ERK1/2 activation in physiologically relevant contexts of the neurohormone action. In addition, we present an overview of the pathophysiological role of the CRH system, which highlights the need for a precise definition of CRHRs signaling at molecular level to identify novel targets for pharmacological intervention in neuroendocrine tissues and specific brain areas involved in CRH-related disorders.
Collapse
Affiliation(s)
- Carolina Inda
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- DFBMCFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia G Armando
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Paula A Dos Santos Claro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- DFBMCFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
47
|
Okada S, Yamaguchi N. Possible role of adrenoceptors in the hypothalamic paraventricular nucleus in corticotropin-releasing factor-induced sympatho-adrenomedullary outflow in rats. Auton Neurosci 2017; 203:74-80. [PMID: 28202248 DOI: 10.1016/j.autneu.2017.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/30/2016] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
AIMS A functional interaction between the corticotropin-releasing factor (CRF) system and noradrenergic neurons in the brain has been suggested. In the present study, we investigated the interrelationship between the central CRF-induced elevation of plasma catecholamines and adrenoceptor activation in the paraventricular nucleus of the hypothalamus (PVN) using urethane-anesthetized rats. MAIN METHODS In rats under urethane anesthesia, a femoral venous line was inserted for infusion of saline, and a femoral arterial line was inserted for collecting blood samples. Next, animals were placed in a stereotaxic apparatus for the application of test agents. Catecholamines in the plasma were extracted by alumina absorption and were assayed with high-performance liquid chromatography with electrochemical detection. Quantification of noradrenaline in rat PVN microdialysates was performed with high-performance liquid chromatography with electrochemical detection. KEY FINDINGS We showed that centrally administered CRF elevated noradrenaline release in the PVN. Furthermore, we demonstrated that microinjection of phenylephrine into the PVN induced elevation of plasma levels of adrenaline, but not of noradrenaline, whereas microinjection of isoproterenol into the PVN induced elevation of plasma levels of noradrenaline, but not of adrenaline. Bilateral blockade of adrenoceptors in the PVN revealed that phentolamine significantly suppressed the CRF-induced elevation of plasma adrenaline level, while propranolol significantly CRF-induced elevation of plasma noradrenaline level. SIGNIFICANCE Our results suggest that centrally administered CRF-induced elevation of plasma levels of adrenaline and noradrenaline can be mediated via activation of α-adrenoceptors and β-adrenoceptors, respectively, in the rat PVN.
Collapse
Affiliation(s)
- Shoshiro Okada
- Department of Pharmacology, Graduate School of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan.
| | - Naoko Yamaguchi
- Department of Pharmacology, Graduate School of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
48
|
Involvement of CRFR 1 in the Basolateral Amygdala in the Immediate Fear Extinction Deficit. eNeuro 2016; 3:eN-NWR-0084-16. [PMID: 27844053 PMCID: PMC5093152 DOI: 10.1523/eneuro.0084-16.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 11/21/2022] Open
Abstract
Several animal and clinical studies have highlighted the ineffectiveness of fear extinction sessions delivered shortly after trauma exposure. This phenomenon, termed the immediate extinction deficit, refers to situations in which extinction programs applied shortly after fear conditioning may result in the reduction of fear behaviors (in rodents, frequently measured as freezing responses to the conditioned cue) during extinction training, but failure to consolidate this reduction in the long term. The molecular mechanisms driving this immediate extinction resistance remain unclear. Here we present evidence for the involvement of the corticotropin releasing factor (CRF) system in the basolateral amygdala (BLA) in male Wistar rats. Intra-BLA microinfusion of the CRFR1 antagonist NBI30775 enhances extinction recall, whereas administration of the CRF agonist CRF6–33 before delayed extinction disrupts recall of extinction. We link the immediate fear extinction deficit with dephosphorylation of GluA1 glutamate receptors at Ser845 and enhanced activity of the protein phosphatase calcineurin in the BLA. Their reversal after treatment with the CRFR1 antagonist indicates their dependence on CRFR1 actions. These findings can have important implications for the improvement of therapeutic approaches to trauma, as well as furthering our understanding of the neurobiological mechanisms underlying fear-related disorders.
Collapse
|
49
|
Henckens MJAG, Deussing JM, Chen A. Region-specific roles of the corticotropin-releasing factor-urocortin system in stress. Nat Rev Neurosci 2016; 17:636-51. [PMID: 27586075 DOI: 10.1038/nrn.2016.94] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dysregulation of the corticotropin-releasing factor (CRF)-urocortin (UCN) system has been implicated in stress-related psychopathologies such as depression and anxiety. It has been proposed that CRF-CRF receptor type 1 (CRFR1) signalling promotes the stress response and anxiety-like behaviour, whereas UCNs and CRFR2 activation mediate stress recovery and the restoration of homeostasis. Recent findings, however, provide clear evidence that this view is overly simplistic. Instead, a more complex picture has emerged that suggests that there are brain region- and cell type-specific effects of CRFR signalling that are influenced by the individual's prior experience and that shape molecular, cellular and ultimately behavioural responses to stressful challenges.
Collapse
Affiliation(s)
- Marloes J A G Henckens
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
50
|
Kennedy BC, Dimova JG, Dakoji S, Yuan LL, Gewirtz JC, Tran PV. Deletion of novel protein TMEM35 alters stress-related functions and impairs long-term memory in mice. Am J Physiol Regul Integr Comp Physiol 2016; 311:R166-78. [PMID: 27170659 DOI: 10.1152/ajpregu.00066.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/04/2016] [Indexed: 12/20/2022]
Abstract
The mounting of appropriate emotional and neuroendocrine responses to environmental stressors critically depends on the hypothalamic-pituitary-adrenal (HPA) axis and associated limbic circuitry. Although its function is currently unknown, the highly evolutionarily conserved transmembrane protein 35 (TMEM35) is prominently expressed in HPA circuitry and limbic areas, including the hippocampus and amygdala. To investigate the possible involvement of this protein in neuroendocrine function, we generated tmem35 knockout (KO) mice to characterize the endocrine, behavioral, electrophysiological, and proteomic alterations caused by deletion of the tmem35 gene. While capable of mounting a normal corticosterone response to restraint stress, KO mice showed elevated basal corticosterone accompanied by increased anxiety-like behavior. The KO mice also displayed impairment of hippocampus-dependent fear and spatial memories. Given the intact memory acquisition but a deficit in memory retention in the KO mice, TMEM35 is likely required for long-term memory consolidation. This conclusion is further supported by a loss of long-term potentiation in the Schaffer collateral-CA1 pathway in the KO mice. To identify putative molecular pathways underlying alterations in plasticity, proteomic analysis of synaptosomal proteins revealed lower levels of postsynaptic molecules important for synaptic plasticity in the KO hippocampus, including PSD95 and N-methyl-d-aspartate receptors. Pathway analysis (Ingenuity Pathway Analysis) of differentially expressed synaptic proteins in tmem35 KO hippocampus implicated molecular networks associated with specific cellular and behavioral functions, including decreased long-term potentiation, and increased startle reactivity and locomotion. Collectively, these data suggest that TMEM35 is a novel factor required for normal activity of the HPA axis and limbic circuitry.
Collapse
Affiliation(s)
- Bruce C Kennedy
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Center for Neurobehavioral Development, University of Minnesota, Minneapolis, Minnesota
| | - Jiva G Dimova
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | - Srikanth Dakoji
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota; and
| | - Li-Lian Yuan
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Jonathan C Gewirtz
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota; Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota; Center for Neurobehavioral Development, University of Minnesota, Minneapolis, Minnesota
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota; and Center for Neurobehavioral Development, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|