1
|
Rye CS, Milton AL. Glutamate receptor expression in the PL-BLA circuit is associated with susceptibility to showing the PTSD-like phenotype. Neurobiol Learn Mem 2025; 219:108051. [PMID: 40157419 DOI: 10.1016/j.nlm.2025.108051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/07/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
While many individuals experience traumatic events during their lifetimes, only some go on to develop post-traumatic stress disorder (PTSD). This susceptibility and resilience to developing PTSD can be modelled in rodents using the stress-enhanced fear learning (SEFL) procedure, in which rats are exposed to a session of massed, unpredictable footshocks and subsequently assessed on tasks of adaptive fear learning. It has previously been observed that subpopulations of rats are susceptible and resilient to showing the PTSD-like phenotype following SEFL, and that these rats show differences in glutamate receptor expression in the basolateral amygdala. However, it is currently unknown whether structural differences are observed in other brain regions implicated in stress responding and memory. Using the refined SEFL procedure, this study aimed to determine whether expression of GluN2B, GluA1 and GluA2 receptor subunits in the prelimbic and infralimbic cortices, and dorsal hippocampus could be correlated to the SEFL-phenotype or shock experience in male rats. Here we show that following SEFL, differences can be observed in receptor subunit expression in the infralimbic cortex and dorsal hippocampus as a function of shock experience, whilst differences in the prelimbic cortex are associated with susceptibility. Importantly, these structural changes can be observed in male rats that are group-housed and exposed to 13-shocks rather than 15-shocks, indicating that the refined SEFL procedure offers a robust animal analogue of the non-associative fear sensitisation that occurs in PTSD. Future studies using this procedure could pave the way to the eventual development of pharmacological treatments to alleviate or prevent stress-induced psychopathology in susceptible individuals.
Collapse
Affiliation(s)
- Charlotte S Rye
- Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, United Kingdom; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom.
| | - Amy L Milton
- Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, United Kingdom
| |
Collapse
|
2
|
Zhang J, Duan J, Li W, Wang X, Ren S, Ye L, Liu F, Tian X, Xie Y, Huang Y, Sun Y, Song N, Li T, Cai X, Liu Z, Zhou H, Huang C, Li Y, Zhu S, Guo F. An antidepressant mechanism underlying the allosteric inhibition of GluN2D-incorporated NMDA receptors at GABAergic interneurons. SCIENCE ADVANCES 2025; 11:eadq0444. [PMID: 40043126 PMCID: PMC11881904 DOI: 10.1126/sciadv.adq0444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025]
Abstract
N-methyl-d-aspartate receptors (NMDARs), key excitatory ion channels, have gained attention as anti-depression targets. NMDARs consist of two GluN1 and two GluN2 subunits (2A-2D), which determine their pharmacological properties. Few compounds selectively targeting GluN2 subunits with antidepressant effects have been identified. Here, we present YY-23, a compound that selectively inhibits GluN2C- or GluN2D-containing NMDARs. Cryo-EM analysis revealed that YY-23 binds to the transmembrane domain of the GluN2D subunit. YY-23 primarily affects GluN2D-containing NMDARs on GABAergic interneurons in the prefrontal cortex, suppressing GABAergic neurotransmission and enhancing excitatory transmission. Behavioral assays demonstrate YY-23's rapid antidepressant effects in both stress-naïve and stress-exposed models, which are lost in mice with global or selective knockout of the grin2d gene in parvalbumin-positive interneurons. These findings highlight GluN2D-containing NMDARs on GABAergic interneurons as potential depression treatment targets.
Collapse
Affiliation(s)
- Jilin Zhang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jinjin Duan
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Wei Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Xian Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Shimin Ren
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Luyu Ye
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fang Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoting Tian
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Xie
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yiming Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yidi Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nan Song
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tianyu Li
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiang Cai
- Oujiang Laboratory, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhiqiang Liu
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| | - Hu Zhou
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| | - Chenggang Huang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Li
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Shujia Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Fei Guo
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Gynecology Hospital of Fudan University, No. 128, Shenyang Rd, Yangpu District, Shanghai 200082, China
| |
Collapse
|
3
|
Judd JM, Peay DN, Kim JL, Smith EA, Donnay ME, Miller J, Klein JP, Nagy EK, Acuña AM, Olive MF, Conrad CD. Inhibition of prefrontal glutamatergic neuron activity during the recovery period following chronic stress disrupts fear memory in male rats: potential role of the infralimbic cortex. Learn Mem 2025; 32:a053957. [PMID: 39824647 PMCID: PMC11801481 DOI: 10.1101/lm.053957.124] [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: 05/31/2024] [Accepted: 11/14/2024] [Indexed: 01/20/2025]
Abstract
Chronic stress typically leads to deficits in fear extinction. However, when a delay occurs from the end of chronic stress and the start of fear conditioning (a "recovery"), rats show improved context-cue discrimination, compared to recently stressed rats or nonstressed rats. The infralimbic cortex (IL) is important for fear extinction and undergoes neuronal remodeling after chronic stress ends, which could drive improved context-cue discrimination. Here, glutamatergic IL neurons of Sprague-Dawley male rats were targeted for inhibition using inhibitory designer receptors exclusively activated by designer drugs (DREADDs) and daily injections of clozapine N-oxide (CNO) during a 21-day recovery period from chronic stress. Histological verification confirmed DREADDs in the IL with some spread to nearby medial prefrontal cortex (PFC) regions. CNO administration was then discontinued before fear conditioning started and behavioral testing thereafter so that behavioral assessments occurred without neuronal inhibition. Fear conditioning involved presenting male rats with three tone-foot shock pairings on 1 day, which was followed by 2 days of 15 tone-alone extinction sessions. Daily and repeated inhibition of mainly IL neurons during the 21-day recovery period did not disrupt fear learning or fear extinction in all groups (controls, stressed rats without a recovery, and stressed rats with a recovery). However, chronically stressed rats given a recovery and with DREADD activation showed impaired spontaneous recovery, indicating a failure to form a tone-foot shock association. The findings show that daily inhibition of mainly IL neurons prior to fear conditioning and extinction depends upon the changes that occur during the recovery period following the end of chronic stress.
Collapse
Affiliation(s)
- Jessica M Judd
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Dylan N Peay
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Jinah L Kim
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Elliot A Smith
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Megan E Donnay
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Joel Miller
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Jean-Paul Klein
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Erin K Nagy
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Amanda M Acuña
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| | - Cheryl D Conrad
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
4
|
Grizzell JA, Clarity TT, Rodriguez RM, Marshall ZQ, Cooper MA. Effects of social dominance and acute social stress on morphology of microglia and structural integrity of the medial prefrontal cortex. Brain Behav Immun 2024; 122:353-367. [PMID: 39187049 PMCID: PMC11402560 DOI: 10.1016/j.bbi.2024.08.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic stress increases activity of the brain's innate immune system and impairs function of the medial prefrontal cortex (mPFC). However, whether acute stress triggers similar neuroimmune mechanisms is poorly understood. Across four studies, we used a Syrian hamster model to investigate whether acute stress drives changes in mPFC microglia in a time-, subregion-, and social status-dependent manner. We found that acute social defeat increased expression of ionized calcium binding adapter molecule 1 (Iba1) in the infralimbic (IL) and prelimbic (PL) and altered the morphology Iba1+ cells 1, 2, and 7 days after social defeat. We also investigated whether acute defeat induced tissue degeneration and reductions of synaptic plasticity 2 days post-defeat. We found that while social defeat increased deposition of cellular debris and reduced synaptophysin immunoreactivity in the PL and IL, treatment with minocycline protected against these cellular changes. Finally, we tested whether a reduced conditioned defeat response in dominant compared to subordinate hamsters was associated with changes in microglia reactivity in the IL and PL. We found that while subordinate hamsters and those without an established dominance relationships showed defeat-induced changes in morphology of Iba1+ cells and cellular degeneration, dominant hamsters showed resistance to these effects of social defeat. Taken together, these findings indicate that acute social defeat alters microglial morphology, increases markers of tissue degradation, and impairs structural integrity in the IL and PL, and that experience winning competitive interactions can specifically protect the IL and reduce stress vulnerability.
Collapse
Affiliation(s)
- J Alex Grizzell
- Neuroscience and Behavioral Biology Program, Emory University, United States; Department of Psychology, University of Tennessee Knoxville, United States; Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Thomas T Clarity
- Department of Psychology, University of Tennessee Knoxville, United States
| | - R Mason Rodriguez
- Department of Psychology, University of Tennessee Knoxville, United States
| | - Zachary Q Marshall
- Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Matthew A Cooper
- Department of Psychology, University of Tennessee Knoxville, United States.
| |
Collapse
|
5
|
Guo F, Zhang B, Shen F, Li Q, Song Y, Li T, Zhang Y, Du W, Li Y, Liu W, Cao H, Zhou X, Zheng Y, Zhu S, Li Y, Liu Z. Sevoflurane acts as an antidepressant by suppression of GluN2D-containing NMDA receptors on interneurons. Br J Pharmacol 2024; 181:3483-3502. [PMID: 38779864 DOI: 10.1111/bph.16420] [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: 04/16/2023] [Revised: 10/18/2023] [Accepted: 11/15/2023] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND AND PURPOSE Sevoflurane, a commonly used inhaled anaesthetic known for its favourable safety profile and rapid onset and offset, has not been thoroughly investigated as a potential treatment for depression. In this study, we reveal the mechanism through which sevoflurane delivers enduring antidepressant effects. EXPERIMENTAL APPROACH To assess the antidepressant effects of sevoflurane, behavioural tests were conducted, along with in vitro and ex vivo whole-cell patch-clamp recordings, to examine the effects on GluN1-GluN2 incorporated N-methyl-d-aspartate (NMDA) receptors (NMDARs) and neuronal circuitry in the medial prefrontal cortex (mPFC). Multiple-channel electrophysiology in freely moving mice was performed to evaluate sevoflurane's effects on neuronal activity, and GluN2D knockout (grin2d-/-) mice were used to confirm the requirement of GluN2D for the antidepressant effects. KEY RESULTS Repeated exposure to subanaesthetic doses of sevoflurane produced sustained antidepressant effects lasting up to 2 weeks. Sevoflurane preferentially inhibited GluN2C- and GluN2D-containing NMDARs, causing a reduction in interneuron activity. In contrast, sevoflurane increased action potentials (AP) firing and decreased spontaneous inhibitory postsynaptic current (sIPSC) in mPFC pyramidal neurons, demonstrating a disinhibitory effect. These effects were absent in grin2d-/- mice, and both pharmacological blockade and genetic knockout of GluN2D abolished sevoflurane's antidepressant actions, suggesting that GluN2D is essential for its antidepressant effect. CONCLUSION AND IMPLICATIONS Sevoflurane directly targets GluN2D, leading to a specific decrease in interneuron activity and subsequent disinhibition of pyramidal neurons, which may underpin its antidepressant effects. Targeting the GluN2D subunit could hold promise as a potential therapeutic strategy for treating depression.
Collapse
Affiliation(s)
- Fei Guo
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Bing Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fuyi Shen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qian Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingcai Song
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianyu Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongmei Zhang
- University of Chinese Academy of Sciences, Beijing, China
| | - Weijia Du
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanxi Li
- Institute for Cognitive Neurodynamics, East China University of Science and Technology, Shanghai, China
| | - Wei Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hang Cao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xianjin Zhou
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yinli Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Shujia Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yang Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhiqiang Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
- Anesthesia and Brain Function Research Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Foster JC, Hodges HR, Beloborodova A, Cohodes EM, Phillips MQ, Anderson E, Fagbenro B, Gee DG. Integrating developmental neuroscience with community-engaged approaches to address mental health outcomes for housing-insecure youth: Implications for research, practice, and policy. Dev Cogn Neurosci 2024; 68:101399. [PMID: 38875770 PMCID: PMC11225708 DOI: 10.1016/j.dcn.2024.101399] [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: 12/30/2023] [Revised: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024] Open
Abstract
One in three children in the United States is exposed to insecure housing conditions, including unaffordable, inconsistent, and unsafe housing. These exposures have detrimental impacts on youth mental health. Delineating the neurobehavioral pathways linking exposure to housing insecurity with children's mental health has the potential to inform interventions and policy. However, in approaching this work, carefully considering the lived experiences of youth and families is essential to translating scientific discovery to improve health outcomes in an equitable and representative way. In the current paper, we provide an introduction to the range of stressful experiences that children may face when exposed to insecure housing conditions. Next, we highlight findings from the early-life stress literature regarding the potential neurobehavioral consequences of insecure housing, focusing on how unpredictability is associated with the neural circuitry supporting cognitive and emotional development. We then delineate how community-engaged research (CEnR) approaches have been leveraged to understand the effects of housing insecurity on mental health, and we propose future research directions that integrate developmental neuroscience research and CEnR approaches to maximize the impact of this work. We conclude by outlining practice and policy recommendations that aim to improve the mental health of children exposed to insecure housing.
Collapse
Affiliation(s)
- Jordan C Foster
- Yale University, Department of Psychology, New Haven, CT, United States.
| | - H R Hodges
- University of Minnesota, Institute of Child Development, Minneapolis, MN, United States
| | - Anna Beloborodova
- Yale University, Department of Psychology, New Haven, CT, United States
| | - Emily M Cohodes
- Yale University, Department of Psychology, New Haven, CT, United States
| | | | | | | | - Dylan G Gee
- Yale University, Department of Psychology, New Haven, CT, United States.
| |
Collapse
|
7
|
Park H, Kuplicki R, Paulus MP, Guinjoan SM. Rumination and Overrecruitment of Cognitive Control Circuits in Depression. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:800-808. [PMID: 38703822 PMCID: PMC11305927 DOI: 10.1016/j.bpsc.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Rumination is associated with greater cognitive dysfunction and treatment resistance in major depressive disorder (MDD), but its underlying neural mechanisms are not well understood. Because rumination is characterized by difficulty in controlling negative thoughts, the current study investigated whether rumination was associated with aberrant cognitive control in the absence of negative emotional information. METHODS Individuals with MDD (n = 176) and healthy control individuals (n = 52) completed the stop signal task with varied stop signal difficulty during functional magnetic resonance imaging. In the task, a longer stop signal asynchrony made stopping difficult (hard stop), whereas a shorter stop signal asynchrony allowed more time for stopping (easy stop). RESULTS In participants with MDD, higher rumination intensity was associated with greater neural activity in response to difficult inhibitory control in the frontoparietal regions. Greater activation for difficult inhibitory control associated with rumination was also positively related to state fear. The imaging results provide compelling evidence for the neural basis of inhibitory control difficulties in individuals with MDD with high rumination. CONCLUSIONS The association between higher rumination intensity and greater neural activity in regions involved in difficult inhibitory control tasks may provide treatment targets for interventions aimed at improving inhibitory control and reducing rumination in this population.
Collapse
Affiliation(s)
- Heekyeong Park
- Laureate Institute for Brain Research, Tulsa, Oklahoma; Department of Psychology, University of North Texas at Dallas, Dallas, Texas.
| | | | - Martin P Paulus
- Laureate Institute for Brain Research, Tulsa, Oklahoma; Oxley College of Health Science, University of Tulsa, Tulsa, Oklahoma
| | - Salvador M Guinjoan
- Laureate Institute for Brain Research, Tulsa, Oklahoma; Department of Psychiatry, Oklahoma State University Health Sciences Center, Tulsa, Oklahoma
| |
Collapse
|
8
|
Reid MA, Whiteman SE, Camden AA, Jeffirs SM, Weathers FW. Prefrontal metabolite alterations in individuals with posttraumatic stress disorder: a 7T magnetic resonance spectroscopy study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603137. [PMID: 39071259 PMCID: PMC11275712 DOI: 10.1101/2024.07.16.603137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Evidence from animal and human studies suggests glutamatergic dysfunction in posttraumatic stress disorder (PTSD). The purpose of this study was to investigate glutamate abnormalities in the dorsolateral prefrontal cortex (DLFPC) of individuals with PTSD using 7T MRS, which has better spectral resolution and signal-to-noise ratio than lower field strengths, thus allowing for better spectral quality and higher sensitivity. We hypothesized that individuals with PTSD would have lower glutamate levels compared to trauma-exposed individuals without PTSD and individuals without trauma exposure. Additionally, we explored potential alterations in other neurometabolites and the relationship between glutamate and psychiatric symptoms. Methods Individuals with PTSD (n=27), trauma-exposed individuals without PTSD (n=27), and individuals without trauma exposure (n=26) underwent 7T MRS to measure glutamate and other neurometabolites in the left DLPFC. The severities of PTSD, depression, anxiety, and dissociation symptoms were assessed. Results We found that glutamate was lower in the PTSD and trauma-exposed groups compared to the group without trauma exposure. Furthermore, N-acetylaspartate (NAA) was lower and lactate was higher in the PTSD group compared to the group without trauma exposure. Glutamate was negatively correlated with depression symptom severity in the PTSD group. Glutamate was not correlated with PTSD symptom severity. Conclusion In this first 7T MRS study of PTSD, we observed altered concentrations of glutamate, NAA, and lactate. Our findings provide evidence for multiple possible pathological processes in individuals with PTSD. High-field MRS offers insight into the neurometabolic alterations associated with PTSD and is a powerful tool to probe trauma- and stress-related neurotransmission and metabolism in vivo.
Collapse
Affiliation(s)
- Meredith A. Reid
- Department of Electrical and Computer Engineering, Auburn University, Auburn, Alabama, USA
- AU Neuroimaging Center, Auburn University, Auburn, Alabama, USA
- Alabama Advanced Imaging Consortium, Auburn, Alabama, USA
| | - Sarah E. Whiteman
- Department of Psychological Sciences, Auburn University, Auburn, Alabama, USA
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Abigail A. Camden
- Department of Psychological Sciences, Auburn University, Auburn, Alabama, USA
| | | | - Frank W. Weathers
- Department of Psychological Sciences, Auburn University, Auburn, Alabama, USA
- National Center for PTSD, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Blanchard DC, Canteras NS. Uncertainty and anxiety: Evolution and neurobiology. Neurosci Biobehav Rev 2024; 162:105732. [PMID: 38797459 DOI: 10.1016/j.neubiorev.2024.105732] [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: 02/06/2024] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Anxiety is a complex phenomenon: Its eliciting stimuli and circumstances, component behaviors, and functional consequences are only slowly coming to be understood. Here, we examine defense systems from field studies; laboratory studies focusing on experimental analyses of behavior; and, the fear conditioning literature, with a focus on the role of uncertainty in promoting an anxiety pattern that involves high rates of stimulus generalization and resistance to extinction. Respectively, these different areas provide information on evolved elicitors of defense (field studies); outline a defense system focused on obtaining information about uncertain threat (ethoexperimental analyses); and, provide a simple, well-researched, easily measured paradigm for analysis of nonassociative stress-enhanced fear conditioning (the SEFL). Results suggest that all of these-each of which is responsive to uncertainty-play multiple and interactive roles in anxiety. Brain system findings for some relevant models are reviewed, with suggestions that further analyses of current models may be capable of providing a great deal of additional information about these complex interactions and their underlying biology.
Collapse
Affiliation(s)
- D Caroline Blanchard
- Pacific Bioscience Research Institute, University of Hawaii, Manoa, USA; Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.
| | - Newton S Canteras
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
10
|
Plas SL, Oleksiak CR, Pitre C, Melton C, Moscarello JM, Maren S. Acute stress yields a sex-dependent facilitation of signaled active avoidance in rats. Neurobiol Stress 2024; 31:100656. [PMID: 38994219 PMCID: PMC11238190 DOI: 10.1016/j.ynstr.2024.100656] [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: 04/27/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating disorder characterized by excessive fear, hypervigilance, and avoidance of thoughts, situations or reminders of the trauma. Among these symptoms, relatively little is known about the etiology of pathological avoidance. Here we sought to determine whether acute stress influences avoidant behavior in adult male and female rats. We used a stress procedure (unsignaled footshock) that is known to induce long-term sensitization of fear and potentiate aversive learning. Rats were submitted to the stress procedure and, one week later, underwent two-way signaled active avoidance conditioning (SAA). In this task, rats learn to prevent an aversive outcome (shock) by performing a shuttling response when exposed to a warning signal (tone). We found that acute stress significantly enhanced SAA acquisition rate in females, but not males. Female rats exhibited significantly greater avoidance responding on the first day of training relative to controls, reaching similar levels of performance by the second day. Males that underwent the stress procedure showed similar rates of acquisition to controls but exhibited resistance to extinction. This was manifest as both elevated avoidance and intertrial responding across extinction days relative to non-stressed controls, an effect that was not observed in females. In a second experiment, acute stress sensitized footshock unconditioned responses in males, not females. However, males and females exhibited similar levels of stress-enhanced fear learning (SEFL), which was expressed as sensitized freezing to a shock-paired context. Together, these results reveal that acute stress facilitates SAA performance in both male and female rats, though the nature of this effect is different in the two sexes. We did not observe sex differences in SEFL, suggesting that the stress-induced sex difference in performance was selective for instrumental avoidance. Future work will elucidate the neurobiological mechanisms underlying the differential effect of stress on instrumental avoidance in male and female rats.
Collapse
Affiliation(s)
- Samantha L. Plas
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
- Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Cecily R. Oleksiak
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
- Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Claire Pitre
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| | - Chance Melton
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| | - Justin M. Moscarello
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
- Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Stephen Maren
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
- Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
11
|
Plas SL, Oleksiak CR, Pitre C, Melton C, Moscarello JM, Maren S. Acute stress yields a sex-dependent facilitation of signaled active avoidance in rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.27.591470. [PMID: 38746268 PMCID: PMC11092500 DOI: 10.1101/2024.04.27.591470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating disorder characterized by excessive fear, hypervigilance, and avoidance of thoughts, situations or reminders of the trauma. Among these symptoms, relatively little is known about the etiology of pathological avoidance. Here we sought to determine whether acute stress influences avoidant behavior in adult male and female rats. We used a stress procedure (unsignaled footshock) that is known to induce long-term sensitization of fear and potentiate aversive learning. Rats were submitted to the stress procedure and, one week later, underwent two-way signaled active avoidance conditioning (SAA). In this task, rats learn to prevent an aversive outcome (shock) by performing a shuttling response when exposed to a warning signal (tone). We found that acute stress significantly enhanced SAA acquisition rate in females, but not males. Female rats exhibited significantly greater avoidance responding on the first day of training relative to controls, reaching similar levels of performance by the second day. Males that underwent the stress procedure showed similar rates of acquisition to controls but exhibited resistance to extinction. This was manifest as both elevated avoidance and intertrial responding across extinction days relative to non-stressed controls, an effect that was not observed in females. In a second experiment, acute stress sensitized footshock unconditioned responses in males, not females. However, males and females exhibited similar levels of stress-enhanced fear learning (SEFL), which was expressed as sensitized freezing to a shock-paired context. Together, these results reveal that acute stress facilitates SAA performance in both male and female rats, though the nature of this effect is different in the two sexes. We did not observe sex differences in SEFL, suggesting that the stress-induced sex difference in performance was selective for instrumental avoidance. Future work will elucidate the neurobiological mechanisms underlying the differential effect of stress on instrumental avoidance in male and female rats.
Collapse
Affiliation(s)
- Samantha L. Plas
- Department of Psychological and Brain Sciences, Texas A&M University, College Station
| | - Cecily R. Oleksiak
- Department of Psychological and Brain Sciences, Texas A&M University, College Station
| | - Claire Pitre
- Department of Psychological and Brain Sciences, Texas A&M University, College Station
| | - Chance Melton
- Department of Psychological and Brain Sciences, Texas A&M University, College Station
| | - Justin M. Moscarello
- Department of Psychological and Brain Sciences, Texas A&M University, College Station
| | - Stephen Maren
- Department of Psychological and Brain Sciences, Texas A&M University, College Station
| |
Collapse
|
12
|
Lazzerini Ospri L, Zhan JJ, Thomsen MB, Wang H, Komal R, Tang Q, Messanvi F, du Hoffmann J, Cravedi K, Chudasama Y, Hattar S, Zhao H. Light affects the prefrontal cortex via intrinsically photosensitive retinal ganglion cells. SCIENCE ADVANCES 2024; 10:eadh9251. [PMID: 38552022 PMCID: PMC10980283 DOI: 10.1126/sciadv.adh9251] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 02/23/2024] [Indexed: 04/01/2024]
Abstract
The ventromedial prefrontal cortex (vmPFC) is a part of the limbic system engaged in the regulation of social, emotional, and cognitive states, which are characteristically impaired in disorders of the brain such as schizophrenia and depression. Here, we show that intrinsically photosensitive retinal ganglion cells (ipRGCs) modulate, through light, the integrity, activity, and function of the vmPFC. This regulatory role, which is independent of circadian and mood alterations, is mediated by an ipRGC-thalamic-corticolimbic pathway. Lack of ipRGC signaling in mice causes dendritic degeneration, dysregulation of genes involved in synaptic plasticity, and depressed neuronal activity in the vmPFC. These alterations primarily undermine the ability of the vmPFC to regulate emotions. Our discovery provides a potential light-dependent mechanism for certain PFC-centric disorders in humans.
Collapse
Affiliation(s)
| | - Jesse J. Zhan
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael B. Thomsen
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hui Wang
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruchi Komal
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qijun Tang
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fany Messanvi
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Johann du Hoffmann
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin Cravedi
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yogita Chudasama
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samer Hattar
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
13
|
Marino N, Bedeschi M, Vaccari ME, Cambiaghi M, Tesei A. Glitches in the brain: the dangerous relationship between radiotherapy and brain fog. Front Cell Neurosci 2024; 18:1328361. [PMID: 38515789 PMCID: PMC10956129 DOI: 10.3389/fncel.2024.1328361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Up to approximately 70% of cancer survivors report persistent deficits in memory, attention, speed of information processing, multi-tasking, and mental health functioning, a series of symptoms known as "brain fog." The severity and duration of such effects can vary depending on age, cancer type, and treatment regimens. In particular, every year, hundreds of thousands of patients worldwide undergo radiotherapy (RT) for primary brain tumors and brain metastases originating from extracranial tumors. Besides its potential benefits in the control of tumor progression, recent studies indicate that RT reprograms the brain tumor microenvironment inducing increased activation of microglia and astrocytes and a consequent general condition of neuroinflammation that in case it becomes chronic could lead to a cognitive decline. Furthermore, radiation can induce endothelium reticulum (ER) stress directly or indirectly by generating reactive oxygen species (ROS) activating compensatory survival signaling pathways in the RT-surviving fraction of healthy neuronal and glial cells. In particular, the anomalous accumulation of misfolding proteins in neuronal cells exposed to radiation as a consequence of excessive activation of unfolded protein response (UPR) could pave the way to neurodegenerative disorders. Moreover, exposure of cells to ionizing radiation was also shown to affect the normal proteasome activity, slowing the degradation rate of misfolded proteins, and further exacerbating ER-stress conditions. This compromises several neuronal functions, with neuronal accumulation of ubiquitinated proteins with a consequent switch from proteasome to immunoproteasome that increases neuroinflammation, a crucial risk factor for neurodegeneration. The etiology of brain fog remains elusive and can arise not only during treatment but can also persist for an extended period after the end of RT. In this review, we will focus on the molecular pathways triggered by radiation therapy affecting cognitive functions and potentially at the origin of so-called "brain fog" symptomatology, with the aim to define novel therapeutic strategies to preserve healthy brain tissue from cognitive decline.
Collapse
Affiliation(s)
- Noemi Marino
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Martina Bedeschi
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Melania Elettra Vaccari
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Marco Cambiaghi
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Anna Tesei
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
14
|
Felix-Ortiz AC, Terrell JM, Gonzalez C, Msengi HD, Boggan MB, Ramos AR, Magalhães G, Burgos-Robles A. Prefrontal Regulation of Safety Learning during Ethologically Relevant Thermal Threat. eNeuro 2024; 11:ENEURO.0140-23.2024. [PMID: 38272673 PMCID: PMC10903390 DOI: 10.1523/eneuro.0140-23.2024] [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: 04/29/2023] [Revised: 01/02/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024] Open
Abstract
Learning and adaptation during sources of threat and safety are critical mechanisms for survival. The prelimbic (PL) and infralimbic (IL) subregions of the medial prefrontal cortex (mPFC) have been broadly implicated in the processing of threat and safety. However, how these regions regulate threat and safety during naturalistic conditions involving thermal challenge still remains elusive. To examine this issue, we developed a novel paradigm in which adult mice learned that a particular zone that was identified with visuospatial cues was associated with either a noxious cold temperature ("threat zone") or a pleasant warm temperature ("safety zone"). This led to the rapid development of avoidance behavior when the zone was paired with cold threat or approach behavior when the zone was paired with warm safety. During a long-term test without further thermal reinforcement, mice continued to exhibit robust avoidance or approach to the zone of interest, indicating that enduring spatial-based memories were formed to represent the thermal threat and thermal safety zones. Optogenetic experiments revealed that neural activity in PL and IL was not essential for establishing the memory for the threat zone. However, PL and IL activity bidirectionally regulated memory formation for the safety zone. While IL activity promoted safety memory during normal conditions, PL activity suppressed safety memory especially after a stress pretreatment. Therefore, a working model is proposed in which balanced activity between PL and IL is favorable for safety memory formation, whereas unbalanced activity between these brain regions is detrimental for safety memory after stress.
Collapse
Affiliation(s)
- Ada C Felix-Ortiz
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
| | - Jaelyn M Terrell
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
| | - Carolina Gonzalez
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
| | - Hope D Msengi
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
| | - Miranda B Boggan
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
| | - Angelica R Ramos
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
| | - Gabrielle Magalhães
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts 02215
| | - Anthony Burgos-Robles
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas 78249
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, Texas 78249
| |
Collapse
|
15
|
Briggs JF, McMullen KM. Retrograde amnesia for the stress-induced impairment of extinction: time-dependent and not so forgotten. Learn Mem 2024; 31:a053895. [PMID: 38286522 PMCID: PMC10903941 DOI: 10.1101/lm.053895.123] [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: 10/16/2023] [Accepted: 12/05/2023] [Indexed: 01/31/2024]
Abstract
We investigated whether retrograde amnesia for the stress-induced impairment of extinction retrieval shares similar characteristics with original acquisition memories. The first experiment demonstrated that cycloheximide administered shortly after a single restraint stress session alleviated the impairment of extinction retrieval but not when administered following a longer delay (i.e., the amnesia for stress is time-dependent). A second experiment showed that the retrograde amnesia for stress could be alleviated by a second brief exposure to the stressor. These results demonstrating that amnesia for stress shares characteristics similar to original memories are explained using a retrieval-based memory integration model of retrograde amnesia.
Collapse
Affiliation(s)
- James F Briggs
- Department of Psychology, Susquehanna University, Selinsgrove, Pennsylvania 17870, USA
| | - Kaitlyn M McMullen
- Department of Psychology, Susquehanna University, Selinsgrove, Pennsylvania 17870, USA
| |
Collapse
|
16
|
Reid MA, Whiteman SE, Camden AA, Jeffirs SM, Weathers FW. Prefrontal Metabolite Alterations in Individuals with Posttraumatic Stress Disorder: A 7T Magnetic Resonance Spectroscopy Study. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2024; 8:24705470241277451. [PMID: 39253023 PMCID: PMC11381574 DOI: 10.1177/24705470241277451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Background Evidence from animal and human studies suggests glutamatergic dysfunction in posttraumatic stress disorder (PTSD). The purpose of this study was to investigate glutamate abnormalities in the dorsolateral prefrontal cortex (DLFPC) of individuals with PTSD using 7T MRS, which has better spectral resolution and signal-to-noise ratio than lower field strengths, thus allowing for better spectral quality and higher sensitivity. We hypothesized that individuals with PTSD would have lower glutamate levels compared to trauma-exposed individuals without PTSD and individuals without trauma exposure. Additionally, we explored potential alterations in other neurometabolites and the relationship between glutamate and psychiatric symptoms. Methods Individuals with PTSD (n = 27), trauma-exposed individuals without PTSD (n = 27), and individuals without trauma exposure (n = 26) underwent 7T MRS to measure glutamate and other neurometabolites in the left DLPFC. The severities of PTSD, depression, anxiety, and dissociation symptoms were assessed. Results We found that glutamate was lower in the PTSD and trauma-exposed groups compared to the group without trauma exposure. Furthermore, N-acetylaspartate (NAA) was lower and lactate was higher in the PTSD group compared to the group without trauma exposure. Glutamate was negatively correlated with depression symptom severity in the PTSD group. Glutamate was not correlated with PTSD symptom severity. Conclusion In this first 7T MRS study of PTSD, we observed altered concentrations of glutamate, NAA, and lactate. Our findings provide evidence for multiple possible pathological processes in individuals with PTSD. High-field MRS offers insight into the neurometabolic alterations associated with PTSD and is a powerful tool to probe trauma- and stress-related neurotransmission and metabolism in vivo.
Collapse
Affiliation(s)
- Meredith A. Reid
- Department of Electrical and Computer Engineering, Auburn University, Auburn, Alabama, USA
- AU Neuroimaging Center, Auburn University, Auburn, Alabama, USA
- Alabama Advanced Imaging Consortium, Auburn, Alabama, USA
| | - Sarah E. Whiteman
- Department of Psychological Sciences, Auburn University, Auburn, Alabama, USA
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Abigail A. Camden
- Department of Psychological Sciences, Auburn University, Auburn, Alabama, USA
| | | | - Frank W. Weathers
- Department of Psychological Sciences, Auburn University, Auburn, Alabama, USA
- National Center for PTSD, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Lazzerini-Ospri L, Zhan JJ, Thomsen MB, Wang H, Messanvi F, du Hoffmann J, Cravedi K, Komal R, Chudasama Y, Zhao H, Hattar S. WITHDRAWN: Light affects the prefrontal cortex via intrinsically photosensitive retinal ganglion cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.556752. [PMID: 37808740 PMCID: PMC10557604 DOI: 10.1101/2023.09.22.556752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
This manuscript has been withdrawn by bioRxiv following a formal request by the NIH Intramural Research Integrity Office owing to lack of author consent.
Collapse
|
18
|
Musazzi L, Mingardi J, Ieraci A, Barbon A, Popoli M. Stress, microRNAs, and stress-related psychiatric disorders: an overview. Mol Psychiatry 2023; 28:4977-4994. [PMID: 37391530 DOI: 10.1038/s41380-023-02139-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/23/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023]
Abstract
Stress is a major risk factor for psychiatric disorders. During and after exposure to stressors, the stress response may have pro- or maladaptive consequences, depending on several factors related to the individual response and nature of the stressor. However, the mechanisms mediating the long-term effects of exposure to stress, which may ultimately lead to the development of stress-related disorders, are still largely unknown. Epigenetic mechanisms have been shown to mediate the effects of the environment on brain gene expression and behavior. MicroRNAs, small non-coding RNAs estimated to control the expression of about 60% of all genes by post-transcriptional regulation, are a fundamental epigenetic mechanism. Many microRNAs are expressed in the brain, where they work as fine-tuners of gene expression, with a key role in the regulation of homeostatic balance, and a likely influence on pro- or maladaptive brain changes. Here we have selected a number of microRNAs, which have been strongly implicated as mediators of the effects of stress in the brain and in the development of stress-related psychiatric disorders. For all of them recent evidence is reported, obtained from rodent stress models, manipulation of microRNAs levels with related behavioral changes, and clinical studies of stress-related psychiatric disorders. Moreover, we have performed a bioinformatic analysis of the predicted brain-expressed target genes of the microRNAs discussed, and found a central role for mechanisms involved in the regulation of synaptic function. The complex regulatory role of microRNAs has suggested their use as biomarkers for diagnosis and treatment response, as well as possible therapeutic drugs. While, microRNA-based diagnostics have registered advancements, particularly in oncology and other fields, and many biotech companies have launched miRNA therapeutics in their development pipeline, the development of microRNA-based tests and drugs for brain disorders is comparatively slower.
Collapse
Affiliation(s)
- Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Jessica Mingardi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Italy
- Molecular Pharmacology, Cellular and Behavioral Physiology; Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.
| |
Collapse
|
19
|
dos-Santos RC, Sweeten BLW, Stelly CE, Tasker JG. The Neuroendocrine Impact of Acute Stress on Synaptic Plasticity. Endocrinology 2023; 164:bqad149. [PMID: 37788632 PMCID: PMC11046011 DOI: 10.1210/endocr/bqad149] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023]
Abstract
Stress induces changes in nervous system function on different signaling levels, from molecular signaling to synaptic transmission to neural circuits to behavior-and on different time scales, from rapid onset and transient to delayed and long-lasting. The principal effectors of stress plasticity are glucocorticoids, steroid hormones that act with a broad range of signaling competency due to the expression of multiple nuclear and membrane receptor subtypes in virtually every tissue of the organism. Glucocorticoid and mineralocorticoid receptors are localized to each of the cellular compartments of the receptor-expressing cells-the membrane, cytosol, and nucleus. In this review, we cover the neuroendocrine effects of stress, focusing mainly on the rapid actions of acute stress-induced glucocorticoids that effect changes in synaptic transmission and neuronal excitability by modulating synaptic and intrinsic neuronal properties via activation of presumed membrane glucocorticoid and mineralocorticoid receptors. We describe the synaptic plasticity that occurs in 4 stress-associated brain structures, the hypothalamus, hippocampus, amygdala, and prefrontal cortex, in response to single or short-term stress exposure. The rapid transformative impact of glucocorticoids makes this stress signal a particularly potent effector of acute neuronal plasticity.
Collapse
Affiliation(s)
- Raoni Conceição dos-Santos
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Brook L W Sweeten
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Claire E Stelly
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
20
|
Jiang LX, Huang GD, Tian YL, Cong RX, Meng X, Wang HL, Zhang C, Yu X. Diminished activation of excitatory neurons in the prelimbic cortex leads to impaired working memory capacity in mice. BMC Biol 2023; 21:171. [PMID: 37568146 PMCID: PMC10416384 DOI: 10.1186/s12915-023-01674-3] [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: 02/09/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Working memory capacity impairment is an early sign of Alzheimer's disease, but the underlying mechanisms remain unclear. Clarifying how working memory capacity is affected will help us better understand the pathological mechanism of Alzheimer's disease. We used the olfactory working memory capacity paradigm to evaluate memory capacity in 3-month-old 5XFAD (an animal model of Alzheimer's disease) mice. Immunofluorescence staining of the prefrontal cortex was performed to detect the number of FOS-positive neurons, calmodulin-dependent protein kinase II-positive neurons, and glutamate decarboxylase-positive neurons in the prelimbic cortex and infralimbic cortex. A chemogenetic method was then used to modulate the inhibition and activation of excitatory neurons in the prelimbic cortex of wild-type and 5XFAD mice and to measure the memory capacity of mice. RESULTS Working memory capacity was significantly diminished in 5XFAD mice compared to littermate wild-type mice. Neuronal activation of the prelimbic cortex, but not the infralimbic cortex, was attenuated in 5XFAD mice performing the olfactory working memory capacity task. Subsequently, the FOS-positive neurons were co-localized with both calmodulin-dependent protein kinase II-positive neurons and glutamate decarboxylase-positive neurons. The results showed that the activation of excitatory neurons in the prelimbic cortex was correlated with working memory capacity in mice. Our results further demonstrate that the chemogenetic inhibition of prelimbic cortex excitatory neurons resulted in reduced working memory capacity in wild-type mice, while the chemogenetic activation of prelimbic cortex excitatory neurons improved the working memory capacity of 5XFAD mice. CONCLUSION The diminished activation of prelimbic cortex excitatory neurons in 5XFAD mice during task performance is associated with reduced working memory capacity, and activation modulation of excitatory neurons by chemogenetic methods can improve memory capacity impairment in 5XFAD mice. These findings may provide a new direction for exploring Alzheimer's disease therapeutic approaches.
Collapse
Affiliation(s)
- Li-Xin Jiang
- Peking University Institute of Mental Health (Sixth Hospital), No.51 Huayuanbei Road, Haidian District, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, 100191, China
- Beijing Municipal Key Laboratory for Translational Research On Diagnosis and Treatment of Dementia, Beijing, 100191, China
| | - Geng-Di Huang
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Mental Health Center, Shenzhen Kangning Hospital, No.77 Zhenbi Road, Pingshan District, Shenzhen, 518118, China
- Affiliated Mental Health Center, Southern University of Science and Technology, No.1088 Xueyuan Avenue, Fuguang Community, Taoyuan Street, Nanshan District, Shenzhen, 518118, China
| | - Yong-Lu Tian
- School of Psychological and Cognitive Sciences, Peking University, No.5 Summer Palace Road, Haidian District, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Ri-Xu Cong
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Xue Meng
- National Center of Gerontology, Beijing Hospital, No.1 Dahua Road, Dongdan, Dongcheng District, Beijing, 100005, China
| | - Hua-Li Wang
- Peking University Institute of Mental Health (Sixth Hospital), No.51 Huayuanbei Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, 100191, China.
- Beijing Municipal Key Laboratory for Translational Research On Diagnosis and Treatment of Dementia, Beijing, 100191, China.
| | - Chen Zhang
- Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You'anmenwai, Fengtai District, Beijing, 100069, China.
| | - Xin Yu
- Peking University Institute of Mental Health (Sixth Hospital), No.51 Huayuanbei Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, 100191, China.
- Beijing Municipal Key Laboratory for Translational Research On Diagnosis and Treatment of Dementia, Beijing, 100191, China.
| |
Collapse
|
21
|
Leo DG, Ozdemir H, Lane DA, Lip GYH, Keller SS, Proietti R. At the heart of the matter: how mental stress and negative emotions affect atrial fibrillation. Front Cardiovasc Med 2023; 10:1171647. [PMID: 37408656 PMCID: PMC10319071 DOI: 10.3389/fcvm.2023.1171647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/07/2023] [Indexed: 07/07/2023] Open
Abstract
Atrial fibrillation (AF) is the most common form of cardiac arrhythmia, affecting 2%-3% of the world's population. Mental and emotional stress, as well as some mental health conditions (e.g., depression) have been shown to significantly impact the heart and have been suggested to act both as independent risk factors and triggers in the onset of AF. In this paper, we review the current literature to examine the role that mental and emotional stress have in the onset of AF and summarise the current knowledge on the interaction between the brain and heart, and the cortical and subcortical pathways involved in the response to stress. Review of the evidence suggests that mental and emotional stress negatively affect the cardiac system, potentially increasing the risk for developing and/or triggering AF. Further studies are required to further understand the cortical and sub-cortical structures involved in the mental stress response and how these interact with the cardiac system, which may help in defining new strategies and interventions to prevent the development of, and improve the management of AF.
Collapse
Affiliation(s)
- Donato Giuseppe Leo
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Hizir Ozdemir
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, United Kingdom
| | - Deirdre A. Lane
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- Danish Center for Clinical Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- Danish Center for Clinical Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Simon S. Keller
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Riccardo Proietti
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
22
|
Kaye AP, Rao MG, Kwan AC, Ressler KJ, Krystal JH. A computational model for learning from repeated traumatic experiences under uncertainty. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2023; 23:894-904. [PMID: 37165181 PMCID: PMC11149767 DOI: 10.3758/s13415-023-01085-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 05/12/2023]
Abstract
Traumatic events can lead to lifelong, inflexible adaptations in threat perception and behavior, which characterize posttraumatic stress disorder (PTSD). This process involves associations between sensory cues and internal states of threat and then generalization of the threat responses to previously neutral cues. However, most formulations neglect adaptations to threat that are not specific to those associations. To incorporate nonassociative responses to threat, we propose a computational theory of PTSD based on adaptation to the frequency of traumatic events by using a reinforcement learning momentum model. Recent threat prediction errors generate momentum that influences subsequent threat perception in novel contexts. This model fits primary data acquired from a mouse model of PTSD, in which unpredictable footshocks in one context accelerate threat learning in a novel context. The theory is consistent with epidemiological data that show that PTSD incidence increases with the number of traumatic events, as well as the disproportionate impact of early life trauma. Because the theory proposes that PTSD relates to the average of recent threat prediction errors rather than the strength of a specific association, it makes novel predictions for the treatment of PTSD.
Collapse
Affiliation(s)
- Alfred P Kaye
- Yale University Department of Psychiatry, New Haven, CT, USA.
- VA National Center for PTSD Clinical Neuroscience Division, West Haven, CT, USA.
| | - Manasa G Rao
- Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - Alex C Kwan
- Yale University Department of Psychiatry, New Haven, CT, USA
- Cornell University Meinig School of Biomedical Engineering, Ithaca, NY, USA
| | - Kerry J Ressler
- McLean Hospital, Division of Depression and Anxiety Disorder, Belmont, MA, USA
- Harvard Medical School, Department of Psychiatry, Boston, MA, USA
| | - John H Krystal
- Yale University Department of Psychiatry, New Haven, CT, USA
- VA National Center for PTSD Clinical Neuroscience Division, West Haven, CT, USA
| |
Collapse
|
23
|
Binette AN, Liu J, Bayer H, Crayton KL, Melissari L, Sweck SO, Maren S. Parvalbumin-Positive Interneurons in the Medial Prefrontal Cortex Regulate Stress-Induced Fear Extinction Impairments in Male and Female Rats. J Neurosci 2023; 43:4162-4173. [PMID: 37127359 PMCID: PMC10255009 DOI: 10.1523/jneurosci.1442-22.2023] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/03/2023] Open
Abstract
Stress has profound effects on fear extinction, a form of learning that is essential to behavioral therapies for trauma-related and stressor-related disorders. Recent work reveals that acute footshock stress reduces medial prefrontal cortex (mPFC) activity that is critical for extinction learning. Reductions in mPFC activity may be mediated by parvalbumin (PV)-containing interneurons via feedforward inhibition imposed by amygdala afferents. To test this hypothesis, footshock stress-induced Fos expression was characterized in PV+ and PV- neurons in the prelimbic (PL) and infralimbic (IL) cortices. Footshock stress increased the proportion of PV+ cells expressing Fos in both male and female rats; this effect was more pronounced in IL compared with PL. To determine whether PV+ interneurons in the mPFC mediate stress-induced extinction impairments, we chemogenetically silenced these neurons before an immediate extinction procedure in PV-Cre rats. Clozapine-N-oxide (CNO) did not affect conditioned freezing during the extinction procedure. However, CNO exacerbated extinction retrieval in both male and female rats with relatively high PL expression of designer receptors exclusively activated by designer drugs (DREADD). In contrast, in rats with relatively high IL DREADD expression, CNO produced a modest facilitation of extinction in the earliest retrieval trials, but in male rats only. Conversely, excitation of IL PV interneurons was sufficient to impair delayed extinction in both male and female rats. Finally, chemogenetic inhibition of IL-projecting amygdala neurons reduced the immediate extinction deficit in male, but not female rats. These results reveal that PV interneurons regulate extinction learning under stress in a sex-dependent manner, and this effect is mediated by amygdaloprefrontal projections.SIGNIFICANCE STATEMENT Stress significantly impairs the memory of fear extinction, a type of learning that is central to behavioral therapies for trauma-based and anxiety-based disorders (e.g., post-traumatic stress disorder). Here we show that acute footshock stress recruits parvalbumin (PV) interneurons in the medial prefrontal cortex (mPFC) of male and female rats. Silencing mPFC PV interneurons or mPFC-projecting amygdala neurons during immediate extinction influenced extinction retrieval in a sex-dependent manner. This work highlights the role for PV-containing mPFC interneurons in stress-induced impairments in extinction learning.
Collapse
Affiliation(s)
- Annalise N Binette
- Department of Psychological & Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3474
| | - Jianfeng Liu
- Department of Psychological & Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3474
| | - Hugo Bayer
- Department of Psychological & Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3474
| | - Kennedi L Crayton
- Department of Psychological & Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3474
| | - Laila Melissari
- Department of Psychological & Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3474
| | - Samantha O Sweck
- Department of Psychological & Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3474
| | - Stephen Maren
- Department of Psychological & Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3474
| |
Collapse
|
24
|
Shin S, Lee S. The impact of environmental factors during maternal separation on the behaviors of adolescent C57BL/6 mice. Front Mol Neurosci 2023; 16:1147951. [PMID: 37293540 PMCID: PMC10244624 DOI: 10.3389/fnmol.2023.1147951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Neonatal maternal separation is a widely used method to construct an early-life stress model in rodents. In this method, pups are separated from their mothers for several hours every day during the first 2 weeks of life, which results in adverse early-life events. It is a known fact that maternal separation can exert a significant impact on the behavior and psychological health, such as anxiety and depression, in adolescent offspring. However, environmental conditions during maternal separation can differ such as the presence of other animals or by placing pups in a different dam. To investigate the differential effects of various conditions of maternal separation on the behavior of adolescent mice, we created the following groups: (1) iMS group: pups were moved to an isolated room with no other adult mice in a nearby cage, (2) eDam group: the pups randomly exchanged their dams, (3) OF group: pups were shifted to another cage with the bedding material containing maternal odor (olfactory stimulation), and (4) MS group: pups were shifted to another vivarium. From postnatal day (PND) 2-20 (i.e., 19 consecutive days), pups were separated from the dam daily for 4 h and exposed to various environments (MS, iMS, eDam, and OF) or were left undisturbed [control (CON) group]. A series of behavioral assessments were conducted to evaluate locomotion, anxiety, recognition, learning, and memory in adolescent offspring. The results showed that neonatal maternal separation led to impaired recognition memory, motor coordination, and motor skill learning across all groups. However, the iMS group exhibited anxiety-like behavior in the elevated plus maze test and enhanced the extinction of fear memory in the auditory fear conditioning test. The OF and eDam groups displayed partially recovered short-term working memory in the Y-maze test but exhibited opposite exploratory behaviors. The OF group spent more time in the center, while the eDam group spent less time. These findings demonstrated that exposure to different environmental conditions during maternal separation causes behavioral alterations in adolescent offspring, providing a potential explanation for the variation in behavioral phenotypes observed in the early-life stress models.
Collapse
|
25
|
On making (and turning adaptive to) maladaptive aversive memories in laboratory rodents. Neurosci Biobehav Rev 2023; 147:105101. [PMID: 36804263 DOI: 10.1016/j.neubiorev.2023.105101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Fear conditioning and avoidance tasks usually elicit adaptive aversive memories. Traumatic memories are more intense, generalized, inflexible, and resistant to attenuation via extinction- and reconsolidation-based strategies. Inducing and assessing these dysfunctional, maladaptive features in the laboratory are crucial to interrogating posttraumatic stress disorder's neurobiology and exploring innovative treatments. Here we analyze over 350 studies addressing this question in adult rats and mice. There is a growing interest in modeling several qualitative and quantitative memory changes by exposing already stressed animals to freezing- and avoidance-related tests or using a relatively high aversive training magnitude. Other options combine aversive/fearful tasks with post-acquisition or post-retrieval administration of one or more drugs provoking neurochemical or epigenetic alterations reported in the trauma aftermath. It is potentially instructive to integrate these procedures and incorporate the measurement of autonomic and endocrine parameters. Factors to consider when defining the organismic and procedural variables, partially neglected aspects (sex-dependent differences and recent vs. remote data comparison) and suggestions for future research (identifying reliable individual risk and treatment-response predictors) are discussed.
Collapse
|
26
|
Aten S, Du Y, Taylor O, Dye C, Collins K, Thomas M, Kiyoshi C, Zhou M. Chronic Stress Impairs the Structure and Function of Astrocyte Networks in an Animal Model of Depression. Neurochem Res 2023; 48:1191-1210. [PMID: 35796915 PMCID: PMC9823156 DOI: 10.1007/s11064-022-03663-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/18/2022] [Indexed: 01/11/2023]
Abstract
Now astrocytes appear to be the key contributors to the pathophysiology of major depression. Evidence in rodents shows that chronic stress is associated with a decreased expression of astrocytic GFAP-immunoreactivity within the cortex in addition to changes in the complexity and length of astrocyte processes. Furthermore, postmortem brains of individuals with depression have revealed a decrease in astrocyte density. Notably, astrocytes are extensively coupled to one another through gap junctions to form a network, or syncytium, and we have previously demonstrated that syncytial isopotentiality is a mechanism by which astrocytes function as an efficient system with respect to brain homeostasis. Interestingly, the question of how astrocyte network function changes following chronic stress is yet to be elucidated. Here, we sought to examine the effects of chronic stress on network-level astrocyte (dys)function. Using a transgenic aldh1l1-eGFP astrocyte reporter mouse, a six-week unpredictable chronic mild stress (UCMS) paradigm as a rodent model of major depression, and immunohistochemical approaches, we show that the morphology of individual astrocytes is altered by chronic stress exposure. Additionally, in astrocyte syncytial isopotentiality measurement, we found that UCMS impairs the syncytial coupling strength of astrocytes within the hippocampus and prefrontal cortex-two brain regions that have been implicated in the regulation of mood. Together, these findings reveal that chronic stress leads to astrocyte atrophy and impaired gap junction coupling, raising the prospect that both individual and network-level astrocyte functionality are important in the etiology of major depression and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sydney Aten
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yixing Du
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Olivia Taylor
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Courtney Dye
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Kelsey Collins
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Matthew Thomas
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Conrad Kiyoshi
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
- Northern Marianas College, Saipan, MP, USA
| | - Min Zhou
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
27
|
Cooper MA, Grizzell JA, Whitten CJ, Burghardt GM. Comparing the ontogeny, neurobiology, and function of social play in hamsters and rats. Neurosci Biobehav Rev 2023; 147:105102. [PMID: 36804399 PMCID: PMC10023430 DOI: 10.1016/j.neubiorev.2023.105102] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Syrian hamsters show complex social play behavior and provide a valuable animal model for delineating the neurobiological mechanisms and functions of social play. In this review, we compare social play behavior of hamsters and rats and underlying neurobiological mechanisms. Juvenile rats play by competing for opportunities to pin one another and attack their partner's neck. A broad set of cortical, limbic, and striatal regions regulate the display of social play in rats. In hamsters, social play is characterized by attacks to the head in early puberty, which gradually transitions to the flanks in late puberty. The transition from juvenile social play to adult hamster aggression corresponds with engagement of neural ensembles controlling aggression. Play deprivation in rats and hamsters alters dendritic morphology in mPFC neurons and impairs flexible, context-dependent behavior in adulthood, which suggests these animals may have converged on a similar function for social play. Overall, dissecting the neurobiology of social play in hamsters and rats can provide a valuable comparative approach for evaluating the function of social play.
Collapse
Affiliation(s)
- Matthew A Cooper
- Department of Psychology, University of Tennessee Knoxville, Knoxville, TN, USA.
| | - J Alex Grizzell
- Neuroscience and Behavioral Biology, Emory University, Atlanta, GA, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Conner J Whitten
- Department of Psychology, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Gordon M Burghardt
- Department of Psychology, University of Tennessee Knoxville, Knoxville, TN, USA; Department of Ecology & Evolutionary Biology, University of Tennessee Knoxville, Knoxville, TN, USA
| |
Collapse
|
28
|
The times they are a-changin': a proposal on how brain flexibility goes beyond the obvious to include the concepts of "upward" and "downward" to neuroplasticity. Mol Psychiatry 2023; 28:977-992. [PMID: 36575306 PMCID: PMC10005965 DOI: 10.1038/s41380-022-01931-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022]
Abstract
Since the brain was found to be somehow flexible, plastic, researchers worldwide have been trying to comprehend its fundamentals to better understand the brain itself, make predictions, disentangle the neurobiology of brain diseases, and finally propose up-to-date treatments. Neuroplasticity is simple as a concept, but extremely complex when it comes to its mechanisms. This review aims to bring to light an aspect about neuroplasticity that is often not given enough attention as it should, the fact that the brain's ability to change would include its ability to disconnect synapses. So, neuronal shrinkage, decrease in spine density or dendritic complexity should be included within the concept of neuroplasticity as part of its mechanisms, not as an impairment of it. To that end, we extensively describe a variety of studies involving topics such as neurodevelopment, aging, stress, memory and homeostatic plasticity to highlight how the weakening and disconnection of synapses organically permeate the brain in so many ways as a good practice of its intrinsic physiology. Therefore, we propose to break down neuroplasticity into two sub-concepts, "upward neuroplasticity" for changes related to synaptic construction and "downward neuroplasticity" for changes related to synaptic deconstruction. With these sub-concepts, neuroplasticity could be better understood from a bigger landscape as a vector in which both directions could be taken for the brain to flexibly adapt to certain demands. Such a paradigm shift would allow a better understanding of the concept of neuroplasticity to avoid any data interpretation bias, once it makes clear that there is no morality with regard to the organic and physiological changes that involve dynamic biological systems as seen in the brain.
Collapse
|
29
|
Bonifacino T, Mingardi J, Facchinetti R, Sala N, Frumento G, Ndoj E, Valenza M, Paoli C, Ieraci A, Torazza C, Balbi M, Guerinoni M, Muhammad N, Russo I, Milanese M, Scuderi C, Barbon A, Steardo L, Bonanno G, Popoli M, Musazzi L. Changes at glutamate tripartite synapses in the prefrontal cortex of a new animal model of resilience/vulnerability to acute stress. Transl Psychiatry 2023; 13:62. [PMID: 36806044 PMCID: PMC9938874 DOI: 10.1038/s41398-023-02366-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/20/2023] Open
Abstract
Stress represents a main risk factor for psychiatric disorders. Whereas it is known that even a single trauma may induce psychiatric disorders in humans, the mechanisms of vulnerability to acute stressors have been little investigated. In this study, we generated a new animal model of resilience/vulnerability to acute footshock (FS) stress in rats and analyzed early functional, molecular, and morphological determinants of stress vulnerability at tripartite glutamate synapses in the prefrontal cortex (PFC). We found that adult male rats subjected to FS can be deemed resilient (FS-R) or vulnerable (FS-V), based on their anhedonic phenotype 24 h after stress exposure, and that these two populations are phenotypically distinguishable up to two weeks afterwards. Basal presynaptic glutamate release was increased in the PFC of FS-V rats, while depolarization-evoked glutamate release and synapsin I phosphorylation at Ser9 were increased in both FS-R and FS-V. In FS-R and FS-V rats the synaptic expression of GluN2A and apical dendritic length of prelimbic PFC layers II-III pyramidal neurons were decreased, while BDNF expression was selectively reduced in FS-V. Depolarization-evoked (carrier-mediated) glutamate release from astroglia perisynaptic processes (gliosomes) was selectively increased in the PFC of FS-V rats, while GLT1 and xCt levels were higher and GS expression reduced in purified PFC gliosomes from FS-R. Overall, we show for the first time that the application of the sucrose intake test to rats exposed to acute FS led to the generation of a novel animal model of resilience/vulnerability to acute stress, which we used to identify early determinants of maladaptive response related to behavioral vulnerability to stress.
Collapse
Affiliation(s)
- Tiziana Bonifacino
- grid.5606.50000 0001 2151 3065Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Jessica Mingardi
- grid.7563.70000 0001 2174 1754School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy ,grid.7637.50000000417571846Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Roberta Facchinetti
- grid.7841.aDepartment of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome, Rome, Italy
| | - Nathalie Sala
- grid.4708.b0000 0004 1757 2822Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Giulia Frumento
- grid.5606.50000 0001 2151 3065Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Elona Ndoj
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marta Valenza
- grid.7841.aDepartment of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome, Rome, Italy
| | - Caterina Paoli
- grid.7563.70000 0001 2174 1754School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy ,grid.5602.10000 0000 9745 6549Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Alessandro Ieraci
- grid.4708.b0000 0004 1757 2822Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy ,grid.449889.00000 0004 5945 6678Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Como, Italy
| | - Carola Torazza
- grid.5606.50000 0001 2151 3065Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Matilde Balbi
- grid.5606.50000 0001 2151 3065Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Michele Guerinoni
- grid.4708.b0000 0004 1757 2822Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Nadeem Muhammad
- grid.5606.50000 0001 2151 3065Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Isabella Russo
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy ,Genetics Unit, IRCCS Istituto Centro S. Giovanni di Dio, Fatebenefratelli, 25125 Brescia, Italy
| | - Marco Milanese
- grid.5606.50000 0001 2151 3065Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy ,grid.410345.70000 0004 1756 7871IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Caterina Scuderi
- grid.7841.aDepartment of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome, Rome, Italy
| | - Alessandro Barbon
- grid.7637.50000000417571846Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca Steardo
- grid.7841.aDepartment of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome, Rome, Italy
| | - Giambattista Bonanno
- grid.5606.50000 0001 2151 3065Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy ,grid.410345.70000 0004 1756 7871IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| |
Collapse
|
30
|
The lifetime impact of stress on fear regulation and cortical function. Neuropharmacology 2023; 224:109367. [PMID: 36464208 DOI: 10.1016/j.neuropharm.2022.109367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
A variety of stressful experiences can influence the ability to form and subsequently inhibit fear memory. While nonsocial stress can impact fear learning and memory throughout the lifespan, psychosocial stressors that involve negative social experiences or changes to the social environment have a disproportionately high impact during adolescence. Here, we review converging lines of evidence that suggest that development of prefrontal cortical circuitry necessary for both social experiences and fear learning is altered by stress exposure in a way that impacts both social and fear behaviors throughout the lifespan. Further, we suggest that psychosocial stress, through its impact on the prefrontal cortex, may be especially detrimental during early developmental periods characterized by higher sociability. This article is part of the Special Issue on 'Fear, Anxiety and PTSD'.
Collapse
|
31
|
Witztum J, Singh A, Zhang R, Johnson M, Liston C. An automated platform for Assessing Working Memory and prefrontal circuit function. Neurobiol Stress 2023; 24:100518. [PMID: 36970451 PMCID: PMC10033752 DOI: 10.1016/j.ynstr.2023.100518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 01/02/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Working memory is a process for actively maintaining and updating task-relevant information, despite interference from competing inputs, and is supported in part by sustained activity in prefrontal cortical pyramidal neurons and coordinated interactions with inhibitory interneurons, which may serve to regulate interference. Chronic stress has potent effects on working memory performance, possibly by interfering with these interactions or by disrupting long-range inputs from key upstream brain regions. Still, the mechanisms by which chronic stress disrupts working memory are not well understood, due in part to a need for scalable, easy-to-implement behavioral assays that are compatible with two-photon calcium imaging and other tools for recording from large populations of neurons. Here, we describe the development and validation of a platform that was designed specifically for automated, high-throughput assessments of working memory and simultaneous two-photon imaging in chronic stress studies. This platform is relatively inexpensive and easy to build; fully automated and scalable such that one investigator can test relatively large cohorts of animals concurrently; fully compatible with two-photon imaging, yet also designed to mitigate head-fixation stress; and can be easily adapted for other behavioral paradigms. Our validation data confirm that mice could be trained to perform a delayed response working memory task with relatively high-fidelity over the course of ∼15 days. Two-photon imaging data validate the feasibility of recording from large populations of cells during working memory tasks performance and characterizing their functional properties. Activity patterns in >70% of medial prefrontal cortical neurons were modulated by at least one task feature, and a majority of cells were engaged by multiple task features. We conclude with a brief literature review of the circuit mechanisms supporting working memory and their disruption in chronic stress states-highlighting directions for future research enabled by this platform.
Collapse
|
32
|
Nishimura KJ, Poulos A, Drew MR, Rajbhandari AK. Know thy SEFL: Fear sensitization and its relevance to stressor-related disorders. Neurosci Biobehav Rev 2022; 142:104884. [PMID: 36174795 DOI: 10.1016/j.neubiorev.2022.104884] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/07/2022] [Accepted: 09/17/2022] [Indexed: 11/27/2022]
Abstract
Extreme stress can cause long-lasting changes in affective behavior manifesting in conditions such as post-traumatic stress disorder (PTSD). Understanding the biological mechanisms that govern trauma-induced behavioral dysregulation requires reliable and rigorous pre-clinical models that recapitulate multiple facets of this complex disease. For decades, Pavlovian fear conditioning has been a dominant paradigm for studying the effects of trauma through an associative learning framework. However, severe stress also causes long-lasting nonassociative fear sensitization, which is often overlooked in Pavlovian fear conditioning studies. This paper synthesizes recent research on the stress-enhanced fear learning (SEFL) paradigm, a valuable rodent model that can dissociate associative and nonassociative effects of stress. We discuss evidence that the SEFL paradigm produces nonassociative fear sensitization that is distinguishable from Pavlovian fear conditioning. We also discuss key biological variables, such as age and sex, neural circuit mechanisms, and crucial gaps in knowledge. We argue that nonassociative fear sensitization deserves more attention within current PTSD models and that SEFL provides a valuable complement to Pavlovian conditioning research on trauma-related pathology.
Collapse
Affiliation(s)
- Kenji J Nishimura
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas, USA, 78712
| | - Andrew Poulos
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, USA, 12222
| | - Michael R Drew
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas, USA, 78712
| | | |
Collapse
|
33
|
Hu W, Zhao X, Liu Y, Ren Y, Wei Z, Tang Z, Tian Y, Sun Y, Yang J. Reward sensitivity modulates the brain reward pathway in stress resilience via the inherent neuroendocrine system. Neurobiol Stress 2022; 20:100485. [PMID: 36132434 PMCID: PMC9483565 DOI: 10.1016/j.ynstr.2022.100485] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/16/2022] [Accepted: 09/02/2022] [Indexed: 12/03/2022] Open
Abstract
In the previous 10 years, researchers have suggested a critical role for the brain reward system in stress resilience. However, no study has provided an empirical link between activity in the mesostriatal reward regions during stress and the recovery of cortisol stress response. Moreover, although reward sensitivity as a trait has been demonstrated to promote stress resilience, it remains unclear whether it modulates the brain reward system in stress resilience and how this effect is achieved by the inherent neuroendocrine system. To investigate these uncertainties, 70 young adults were recruited to participate in a ScanSTRESS task, and their brain imaging data and saliva samples (for cortisol assay) were collected during the task. In addition, we assessed reward sensitivity, cortisol awakening response, and intrinsic functional connectivity of the brain in all the participants. We found that left putamen activation during stress exposure positively predicted cortisol recovery. In addition, reward sensitivity was positively linked with activation of the left putamen, and this relationship was serially mediated by the cortisol awakening response and right hippocampus-left inferior frontal gyrus intrinsic connectivity. These findings suggest that reward sensitivity modulates reward pathways in stress resilience through the interplay of the diurnal stress response system and network of the hippocampus-prefrontal circuitry. Summarily, the current study built a model to highlight the dynamic and multifaceted interaction between pertinent allostatic factors in the reward-resilience pathway and uncovered new insight into the resilience function of the mesostriatal reward system during stress. Cortisol recovery can be predicted by activation of the left putamen in stress. Activation of the left putamen was positively linked with reward sensitivity. This relationship was serially mediated by the cortisol awakening response and right hippocampus-left inferior frontal gyrus intrinsic coupling.
Collapse
Affiliation(s)
- Weiyu Hu
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| | - Xiaolin Zhao
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| | - Yadong Liu
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| | - Yipeng Ren
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| | - Zhenni Wei
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| | - Zihan Tang
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| | - Yun Tian
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| | - Yadong Sun
- Faculty of Psychology, Southwest University, Chongqing, 400715, China
| | - Juan Yang
- Faculty of Psychology, Southwest University, Chongqing, 400715, China.,Key Laboratory of Cognition and Personality, Southwest University, Chongqing, 400715, China
| |
Collapse
|
34
|
Yoshida M, Hasegawa S, Taniguchi M, Mouri A, Suzuki C, Yoshimi A, Mamiya T, Ozaki N, Noda Y. Memantine ameliorates the impairment of social behaviors induced by a single social defeat stress as juveniles. Neuropharmacology 2022; 217:109208. [PMID: 35926580 DOI: 10.1016/j.neuropharm.2022.109208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022]
Abstract
Clinically, juveniles are more sensitive to stress than adults, and exposure to stress as juveniles prolongs psychiatric symptoms and causes treatment resistance. However, the efficacy of antidepressants for juveniles with psychiatric disorders is unknown. In the present study, we investigated whether the expression or development of impaired social behavior was attenuated by memantine, a NMDA receptor antagonist. In addition, we clarified the molecular mechanisms related to intracellular signal transduction through NMDA receptors and the ameliorating effect of memantine in mice with impaired social behavior. Acute administration of memantine before the social interaction test, but not before exposure to social defeat stress, attenuated social behavioral impairment. A single social defeat stress increased the phosphorylation of NMDA receptor subunit GluN2A and extracellular-signal-related kinase 1/2 (ERK1/2). Memantine inhibited the increase of phosphorylated GluN2A and ERK1/2 resulting from social interaction behavior. In both GluN2A deficient and pharmacological blockaded mice, social behavioral impairment was not observed in the social interaction test through regulation of ERK1/2 phosphorylation. These findings suggest that memantine ameliorates social behavioral impairment in mice exposed to a single social defeat stress as juveniles by regulating the NMDA receptor and subsequent ERK1/2 signaling activation. Memantine may constitute a novel therapeutic drug for stress-related psychiatric disorders in juveniles with adverse juvenile experiences.
Collapse
Affiliation(s)
- Mikio Yoshida
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Sho Hasegawa
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Masayuki Taniguchi
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Akihiro Mouri
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Chiharu Suzuki
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Akira Yoshimi
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Takayoshi Mamiya
- Department of Chemical Pharmacology, Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Noda
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan.
| |
Collapse
|
35
|
Bernardo A, Lee P, Marcotte M, Mian MY, Rezvanian S, Sharmin D, Kovačević A, Savić MM, Cook JM, Sibille E, Prevot TD. Symptomatic and neurotrophic effects of GABAA receptor positive allosteric modulation in a mouse model of chronic stress. Neuropsychopharmacology 2022; 47:1608-1619. [PMID: 35701547 PMCID: PMC9283409 DOI: 10.1038/s41386-022-01360-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 12/27/2022]
Abstract
Chronic stress is a risk factor for Major Depressive Disorder (MDD), and in rodents, it recapitulates human behavioral, cellular and molecular changes. In MDD and after chronic stress, neuronal dysfunctions and deficits in GABAergic signaling are observed and responsible for symptom severity. GABA signals predominantly through GABAA receptors (GABAA-R) composed of various subunit types that relate to downstream outcomes. Activity at α2-GABAA-Rs contributes to anxiolytic properties, α5-GABAA-Rs to cognitive functions, and α1-GABAA-Rs to sedation. Therefore, a therapy aiming at increasing α2- and α5-GABAA-Rs activity, but devoid of α1-GABAA-R activity, has potential to address several symptomologies of depression while avoiding side-effects. This study investigated the activity profiles and behavioral efficacy of two enantiomers of each other (GL-II-73 and GL-I-54), separately and as a racemic mixture (GL-RM), and potential disease-modifying effects on neuronal morphology. Results confirm GL-I-54 and GL-II-73 exert positive allosteric modulation at the α2-, α3-, α5-GABAA-Rs and α5-containing GABAA-Rs, respectively, and separately reduces immobility in the forced swim test and improves stress-induced spatial working memory deficits. Using unpredictable chronic mild stress (UCMS), we show that acute and chronic administration of GL-RM provide pro-cognitive effects, with mild efficacy on mood symptoms, although at lower doses avoiding sedation. Morphology studies showed reversal of spine density loss caused by UCMS after chronic GL-RM treatment at apical and basal dendrites of the PFC and CA1. Together, these results support using a racemic mixture with combined α2-, α3-, α5-GABAA-R profile to reverse chronic stress-induced mood symptoms, cognitive deficits, and with anti-stress neurotrophic effects.
Collapse
Affiliation(s)
- Ashley Bernardo
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada
| | - Philip Lee
- grid.17063.330000 0001 2157 2938Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Michael Marcotte
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada
| | - Md Yeunus Mian
- grid.267468.90000 0001 0695 7223Department of Chemistry and Biochemistry, University of Wisconsin–Milwaukee, Milwaukee, USA
| | - Sepideh Rezvanian
- grid.267468.90000 0001 0695 7223Department of Chemistry and Biochemistry, University of Wisconsin–Milwaukee, Milwaukee, USA
| | - Dishary Sharmin
- grid.267468.90000 0001 0695 7223Department of Chemistry and Biochemistry, University of Wisconsin–Milwaukee, Milwaukee, USA
| | - Aleksandra Kovačević
- grid.7149.b0000 0001 2166 9385Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Miroslav M. Savić
- grid.7149.b0000 0001 2166 9385Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - James M. Cook
- grid.267468.90000 0001 0695 7223Department of Chemistry and Biochemistry, University of Wisconsin–Milwaukee, Milwaukee, USA
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada. .,Department of Psychiatry, University of Toronto, Toronto, Canada.
| | - Thomas D. Prevot
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
36
|
López-Terrones E, Celada P, Riga MS, Artigas F. Preferential in vivo inhibitory action of serotonin in rat infralimbic versus prelimbic cortex: relevance for antidepressant treatments. Cereb Cortex 2022; 32:3000-3013. [DOI: 10.1093/cercor/bhab396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/14/2022] Open
Abstract
Abstract
The infralimbic (IL) cortex is the rodent equivalent of human ventral anterior cingulate cortex (vACC), which plays a key role in the pathophysiology and treatment of major depressive disorder (MDD). The modulation of glutamatergic neurotransmission in IL [but not in the adjacent prelimbic (PrL) cortex] evokes antidepressant-like or depressive-like behaviors, associated with changes in serotonin (5-HT) function, highlighting the relevance of glutamate/serotonin interactions in IL for emotional control. 5-HT modulates neuronal activity in PrL and cingulate (Cg) cortex but its effects in IL are largely unknown. We therefore compared the in vivo effects of 5-HT on pyramidal neuron activity in IL (n = 61) and PrL (n = 50) of anesthetized rats. IL pyramidal neurons were more responsive to physiological dorsal raphe stimulation (0.9 Hz) than PrL neurons (84% vs. 64%, respectively) and were inhibited to a greater extent (64% vs. 36%, respectively). Orthodromic activations (8% in PrL) were absent in IL, whereas biphasic responses were similar (20%) in both areas. Excitations were mediated by 5-HT2A-R activation, whereas inhibitions involved 3 different components: 5-HT1A-R, 5-HT3-R and GABAA-R, respectively. The remarkable inhibitory action of 5-HT in IL suggests that 5-HT-enhancing drugs may exert their antidepressant action by normalizing a glutamatergic hyperactivity in the vACC of MDD patients.
Collapse
Affiliation(s)
- Elena López-Terrones
- Depart. de Neurociències i Terapèutica Experimental , Institut d'Investigacions Biomèdiques de Barcelona, IIBB-CSIC; 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) , Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pau Celada
- Depart. de Neurociències i Terapèutica Experimental , Institut d'Investigacions Biomèdiques de Barcelona, IIBB-CSIC; 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) , Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maurizio S Riga
- Depart. de Neurociències i Terapèutica Experimental , Institut d'Investigacions Biomèdiques de Barcelona, IIBB-CSIC; 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) , Instituto de Salud Carlos III, 28029 Madrid, Spain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER-CSIC) , 41092 Sevilla, Spain
| | - Francesc Artigas
- Depart. de Neurociències i Terapèutica Experimental , Institut d'Investigacions Biomèdiques de Barcelona, IIBB-CSIC; 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) , Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
37
|
Csabai D, Sebők-Tornai A, Wiborg O, Czéh B. A Preliminary Quantitative Electron Microscopic Analysis Reveals Reduced Number of Mitochondria in the Infralimbic Cortex of Rats Exposed to Chronic Mild Stress. Front Behav Neurosci 2022; 16:885849. [PMID: 35600987 PMCID: PMC9115382 DOI: 10.3389/fnbeh.2022.885849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
Exposure to severe, uncontrollable and long-lasting stress is a strong risk factor for the development of numerous mental and somatic disorders. Animal studies document that chronic stress can alter neuronal morphology and functioning in limbic brain structures such as the prefrontal cortex. Mitochondria are intracellular powerhouses generating chemical energy for biochemical reactions of the cell. Recent findings document that chronic stress can lead to changes in mitochondrial function and metabolism. Here, we studied putative mitochondrial damage in response to chronic stress in neurons of the medial prefrontal cortex. We performed a systematic quantitative ultrastructural analysis to examine the consequences of 9-weeks of chronic mild stress on mitochondria number and morphology in the infralimbic cortex of adult male rats. In this preliminary study, we analyzed 4,250 electron microscopic images and 67000 mitochondria were counted and examined in the brains of 4 control and 4 stressed rats. We found significantly reduced number of mitochondria in the infralimbic cortex of the stressed animals, but we could not detect any significant alteration in mitochondrial morphology. These data support the concept that prolonged stress can lead to mitochondrial loss. This in turn may result in impaired energy production. Reduced cellular energy may sensitize the neurons to additional injuries and may eventually trigger the development of psychopathologies.
Collapse
Affiliation(s)
- Dávid Csabai
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Abigél Sebők-Tornai
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
38
|
Maren S. Unrelenting Fear Under Stress: Neural Circuits and Mechanisms for the Immediate Extinction Deficit. Front Syst Neurosci 2022; 16:888461. [PMID: 35520882 PMCID: PMC9062589 DOI: 10.3389/fnsys.2022.888461] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Therapeutic interventions for disorders of fear and anxiety rely on behavioral approaches that reduce pathological fear memories. For example, learning that threat-predictive stimuli are no longer associated with aversive outcomes is central to the extinction of conditioned fear responses. Unfortunately, fear memories are durable, long-lasting, and resistant to extinction, particularly under high levels of stress. This is illustrated by the "immediate extinction deficit," which is characterized by a poor long-term reduction of conditioned fear when extinction procedures are attempted within hours of fear conditioning. Here, I will review recent work that has provided new insight into the neural mechanisms underlying resistance to fear extinction. Emerging studies reveal that locus coeruleus norepinephrine modulates amygdala-prefrontal cortical circuits that are critical for extinction learning. These data suggest that stress-induced activation of brain neuromodulatory systems biases fear memory at the expense of extinction learning. Behavioral and pharmacological strategies to reduce stress in patients undergoing exposure therapy might improve therapeutic outcomes.
Collapse
Affiliation(s)
- Stephen Maren
- Department of Psychological and Brain Sciences, Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
39
|
Namkung H, Thomas KL, Hall J, Sawa A. Parsing neural circuits of fear learning and extinction across basic and clinical neuroscience: Towards better translation. Neurosci Biobehav Rev 2022; 134:104502. [PMID: 34921863 DOI: 10.1016/j.neubiorev.2021.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022]
Abstract
Over the past decades, studies of fear learning and extinction have advanced our understanding of the neurobiology of threat and safety learning. Animal studies can provide mechanistic/causal insights into human brain regions and their functional connectivity involved in fear learning and extinction. Findings in humans, conversely, may further enrich our understanding of neural circuits in animals by providing macroscopic insights at the level of brain-wide networks. Nevertheless, there is still much room for improvement in translation between basic and clinical research on fear learning and extinction. Through the lens of neural circuits, in this article, we aim to review the current knowledge of fear learning and extinction in both animals and humans, and to propose strategies to fill in the current knowledge gap for the purpose of enhancing clinical benefits.
Collapse
Affiliation(s)
- Ho Namkung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK; School of Biosciences, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK
| | - Akira Sawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21287, USA.
| |
Collapse
|
40
|
Reckweg JT, Uthaug MV, Szabo A, Davis AK, Lancelotta R, Mason NL, Ramaekers JG. The clinical pharmacology and potential therapeutic applications of 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT). J Neurochem 2022; 162:128-146. [PMID: 35149998 PMCID: PMC9314805 DOI: 10.1111/jnc.15587] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 11/26/2022]
Abstract
5‐methoxy‐N,N‐dimethyltryptamine (5‐MeO‐DMT) is a naturally occurring tryptamine that primarily acts as an agonist at the 5‐HT1A and 5‐HT2A receptors, whereby affinity for the 5‐HT1A subtype is highest. Subjective effects following 5‐MeO‐DMT administration include distortions in auditory and time perception, amplification of emotional states, and feelings of ego dissolution that usually are short‐lasting, depending on the route of administration. Individual dose escalation of 5‐MeO‐DMT reliably induces a “peak” experience, a state thought to be a core predictor of the therapeutic efficacy of psychedelics. Observational studies and surveys have suggested that single exposure to 5‐MeO‐DMT can cause rapid and sustained reductions in symptoms of depression, anxiety, and stress. 5‐MeO‐DMT also stimulates neuroendocrine function, immunoregulation, and anti‐inflammatory processes, which may contribute to changes in mental health outcomes. To date, only one clinical trial has been published on 5‐MeO‐DMT, demonstrating the safety of vaporized dosing up to 18 mg. Importantly, the rapid onset and short duration of the 5‐MeO‐DMT experience may render it more suitable for individual dose‐finding strategies compared with longer‐acting psychedelics. A range of biotech companies has shown an interest in the development of 5‐MeO‐DMT formulations for a range of medical indications, most notably depression. Commercial development will therefore be the most important resource for bringing 5‐MeO‐DMT to the clinic. However, fundamental research will also be needed to increase understanding of the neurophysiological and neural mechanisms that contribute to the potential clinical effects of 5‐MeO‐DMT and its sustainability and dissemination over time. Such studies are less likely to be conducted as part of drug development programs and are more likely to rely on independent, academic initiatives.
Collapse
Affiliation(s)
- J T Reckweg
- Faculty of Psychology and Neuroscience, Dept of Neuropsychology and psychopharmacology, Maastricht University, The Netherlands
| | - M V Uthaug
- Faculty of Psychology and Neuroscience, Dept of Neuropsychology and psychopharmacology, Maastricht University, The Netherlands
| | - A Szabo
- Norwegian Centre for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.,KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - A K Davis
- Center for Psychedelic Drug Research and Education, College of Social Work, The Ohio State University, Columbus, OH, USA.,Center for Psychedelic and Consciousness Research, Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - R Lancelotta
- Center for Psychedelic Drug Research and Education, College of Social Work, The Ohio State University, Columbus, OH, USA
| | - N L Mason
- Faculty of Psychology and Neuroscience, Dept of Neuropsychology and psychopharmacology, Maastricht University, The Netherlands
| | - J G Ramaekers
- Faculty of Psychology and Neuroscience, Dept of Neuropsychology and psychopharmacology, Maastricht University, The Netherlands
| |
Collapse
|
41
|
Sanacora G, Yan Z, Popoli M. The stressed synapse 2.0: pathophysiological mechanisms in stress-related neuropsychiatric disorders. Nat Rev Neurosci 2022; 23:86-103. [PMID: 34893785 DOI: 10.1038/s41583-021-00540-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 12/25/2022]
Abstract
Stress is a primary risk factor for several neuropsychiatric disorders. Evidence from preclinical models and clinical studies of depression have revealed an array of structural and functional maladaptive changes, whereby adverse environmental factors shape the brain. These changes, observed from the molecular and transcriptional levels through to large-scale brain networks, to the behaviours reveal a complex matrix of interrelated pathophysiological processes that differ between sexes, providing insight into the potential underpinnings of the sex bias of neuropsychiatric disorders. Although many preclinical studies use chronic stress protocols, long-term changes are also induced by acute exposure to traumatic stress, opening a path to identify determinants of resilient versus susceptible responses to both acute and chronic stress. Epigenetic regulation of gene expression has emerged as a key player underlying the persistent impact of stress on the brain. Indeed, histone modification, DNA methylation and microRNAs are closely involved in many aspects of the stress response and reveal the glutamate system as a key player. The success of ketamine has stimulated a whole line of research and development on drugs directly or indirectly targeting glutamate function. However, the challenge of translating the emerging understanding of stress pathophysiology into effective clinical treatments remains a major challenge.
Collapse
Affiliation(s)
- Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Department of Pharmaceutical Sciences, University of Milano, Milan, Italy.
| |
Collapse
|
42
|
Ball KT, Bennardo GM, Roe J, Wunderlich KJ. Dopamine D 1-like receptors in prelimbic, but not infralimbic, medial prefrontal cortex contribute to chronic stress-induced increases in cue-induced relapse to palatable food seeking during forced abstinence. Behav Brain Res 2022; 417:113583. [PMID: 34530043 PMCID: PMC8578442 DOI: 10.1016/j.bbr.2021.113583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/20/2021] [Accepted: 09/09/2021] [Indexed: 01/26/2023]
Abstract
Chronic stress exposure causes increased vulnerability to future relapse-like behavior in male, but not female, rats with a history of palatable food self-administration. These effects are mediated by dopamine D1-like receptors, but the anatomical location of chronic stress' dopaminergic mechanism is not known. Thus, male rats were trained to respond for palatable food pellets in daily sessions. During subsequent forced abstinence from food self-administration, stress was manipulated (0 or 3 h restraint/day for 7 days). Rats also received bilateral microinjections of the D1-like receptor antagonist SCH-23390 (0.25 μg/0.5 μl/side) or vehicle (0.5 μl/side) delivered to either prelimbic or infralimbic medial prefrontal cortex prior to daily treatments. Relapse tests in the presence of food-associated cues were conducted 7 days after the last treatment. Stress caused an increase and a decrease in responding during relapse tests in rats that received prelimbic vehicle and SCH-23390 infusions, respectively, relative to unstressed rats. In rats receiving IL infusions, however, stress caused an increase in responding regardless of whether the infusion was vehicle or SCH-23390. These results establish a specific role for prelimbic D1-like receptors in chronic stress-potentiated relapse.
Collapse
Affiliation(s)
- Kevin T Ball
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA 17815, USA.
| | - Guy M Bennardo
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA 17815, USA
| | - Jonathan Roe
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA 17815, USA
| | - Kyle J Wunderlich
- Department of Psychology, Bloomsburg University of Pennsylvania, 400 E. 2nd St., Bloomsburg, PA 17815, USA
| |
Collapse
|
43
|
Abstract
During evolution, the cerebral cortex advances by increasing in surface and the introduction of new cytoarchitectonic areas among which the prefrontal cortex (PFC) is considered to be the substrate of highest cognitive functions. Although neurons of the PFC are generated before birth, the differentiation of its neurons and development of synaptic connections in humans extend to the 3rd decade of life. During this period, synapses as well as neurotransmitter systems including their receptors and transporters, are initially overproduced followed by selective elimination. Advanced methods applied to human and animal models, enable investigation of the cellular mechanisms and role of specific genes, non-coding regulatory elements and signaling molecules in control of prefrontal neuronal production and phenotypic fate, as well as neuronal migration to establish layering of the PFC. Likewise, various genetic approaches in combination with functional assays and immunohistochemical and imaging methods reveal roles of neurotransmitter systems during maturation of the PFC. Disruption, or even a slight slowing of the rate of neuronal production, migration and synaptogenesis by genetic or environmental factors, can induce gross as well as subtle changes that eventually can lead to cognitive impairment. An understanding of the development and evolution of the PFC provide insight into the pathogenesis and treatment of congenital neuropsychiatric diseases as well as idiopathic developmental disorders that cause intellectual disabilities.
Collapse
Affiliation(s)
- Sharon M Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands.
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, USA.
| |
Collapse
|
44
|
Alexandra Kredlow M, Fenster RJ, Laurent ES, Ressler KJ, Phelps EA. Prefrontal cortex, amygdala, and threat processing: implications for PTSD. Neuropsychopharmacology 2022; 47:247-259. [PMID: 34545196 PMCID: PMC8617299 DOI: 10.1038/s41386-021-01155-7] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 02/08/2023]
Abstract
Posttraumatic stress disorder can be viewed as a disorder of fear dysregulation. An abundance of research suggests that the prefrontal cortex is central to fear processing-that is, how fears are acquired and strategies to regulate or diminish fear responses. The current review covers foundational research on threat or fear acquisition and extinction in nonhuman animals, healthy humans, and patients with posttraumatic stress disorder, through the lens of the involvement of the prefrontal cortex in these processes. Research harnessing advances in technology to further probe the role of the prefrontal cortex in these processes, such as the use of optogenetics in rodents and brain stimulation in humans, will be highlighted, as well other fear regulation approaches that are relevant to the treatment of posttraumatic stress disorder and involve the prefrontal cortex, namely cognitive regulation and avoidance/active coping. Despite the large body of translational research, many questions remain unanswered and posttraumatic stress disorder remains difficult to treat. We conclude by outlining future research directions related to the role of the prefrontal cortex in fear processing and implications for the treatment of posttraumatic stress disorder.
Collapse
Affiliation(s)
- M. Alexandra Kredlow
- grid.38142.3c000000041936754XDepartment of Psychology, Harvard University, Cambridge, MA USA
| | - Robert J. Fenster
- grid.38142.3c000000041936754XDivision of Depression and Anxiety, McLean Hospital; Department of Psychiatry, Harvard Medical School, Cambridge, MA USA
| | - Emma S. Laurent
- grid.38142.3c000000041936754XDepartment of Psychology, Harvard University, Cambridge, MA USA
| | - Kerry J. Ressler
- grid.38142.3c000000041936754XDivision of Depression and Anxiety, McLean Hospital; Department of Psychiatry, Harvard Medical School, Cambridge, MA USA
| | - Elizabeth A. Phelps
- grid.38142.3c000000041936754XDepartment of Psychology, Harvard University, Cambridge, MA USA
| |
Collapse
|
45
|
López-Arnau R, Camarasa J, Carbó ML, Nadal-Gratacós N, Puigseslloses P, Espinosa-Velasco M, Urquizu E, Escubedo E, Pubill D. 3,4-Methylenedioxy methamphetamine, synthetic cathinones and psychedelics: From recreational to novel psychotherapeutic drugs. Front Psychiatry 2022; 13:990405. [PMID: 36262632 PMCID: PMC9574023 DOI: 10.3389/fpsyt.2022.990405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
The utility of classical drugs used to treat psychiatric disorders (e.g., antidepressants, anxiolytics) is often limited by issues of lack of efficacy, delayed onset of action or side effects. Psychoactive substances have a long history of being used as tools to alter consciousness and as a gateway to approach the unknown and the divinities. These substances were initially obtained from plants and animals and more recently by chemical synthesis, and its consumption evolved toward a more recreational use, leading to drug abuse-related disorders, trafficking, and subsequent banning by the authorities. However, these substances, by modulation of certain neurochemical pathways, have been proven to have a beneficial effect on some psychiatric disorders. This evidence obtained under medically controlled conditions and often associated with psychotherapy, makes these substances an alternative to conventional medicines, to which in many cases the patient does not respond properly. Such disorders include post-traumatic stress disease and treatment-resistant depression, for which classical drugs such as MDMA, ketamine, psilocybin and LSD, among others, have already been clinically tested, reporting successful outcomes. The irruption of new psychoactive substances (NPS), especially during the last decade and despite their recreational and illicit uses, has enlarged the library of substances with potential utility on these disorders. In fact, many of them were synthetized with therapeutic purposes and were withdrawn for concrete reasons (e.g., adverse effects, improper pharmacological profile). In this review we focus on the basis, existing evidence and possible use of synthetic cathinones and psychedelics (specially tryptamines) for the treatment of mental illnesses and the properties that should be found in NPS to obtain new therapeutic compounds.
Collapse
Affiliation(s)
- Raúl López-Arnau
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Jordi Camarasa
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Marcel Lí Carbó
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Núria Nadal-Gratacós
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Pharmaceutical Chemistry Group (GQF), IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - Pol Puigseslloses
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - María Espinosa-Velasco
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Edurne Urquizu
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Elena Escubedo
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - David Pubill
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| |
Collapse
|
46
|
Fiedler D, Pape HC, Lange MD. Stress-induced impairment of fear extinction recall is associated with changes in neuronal activity patterns in PVT. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110338. [PMID: 33915218 DOI: 10.1016/j.pnpbp.2021.110338] [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: 09/16/2020] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 10/21/2022]
Abstract
Treatment resistance of anxiety-related disorders often arises from an inappropriate fear expression, impairment in fear extinction, and spontaneous return of fear. Stress exposure is considered a high risk factor for neuropsychiatric disorders, but understanding of the long-term consequences of stress is limited, particularly when it comes to treatment outcome. Therefore, studying the consequences of acute stress would provide critical information on the role of stress in psychopathology. In the present study, we investigated the effect of acute immobilization stress on anxiety-like behavior and on conditioned fear memory. Our results demonstrate that prior stress exposure had no effect on anxiety-related behavior, fear acquisition, as well as fear extinction compared to non-stressed controls, but resulted in significantly higher rates of freezing during recall of extinction, indicating a consolidation failure. Further, immunohistochemical analysis of the expression of the immediate early gene c-Fos after recall of extinction revealed increased neuronal activity in the posterior paraventricular nucleus of the thalamus (PVT) in previously stressed animals compared to non-stressed controls. These results indicate, firstly, that acute stress affects long-term fear memory even after successful extinction training, and secondly, a strong involvement of the PVT in maladaptive fear responses induced by prior stress. Thus, stress-induced changes in PVT neuronal activity might be of importance for the pathophysiology of stress-sensitive anxiety-related psychiatric disorders, since exposure to an earlier acute stressor could counteract the success of therapy.
Collapse
Affiliation(s)
- D Fiedler
- Institute of Physiology I, Westfälische Wilhelms-University Münster, 48149 Münster, Germany
| | - H C Pape
- Institute of Physiology I, Westfälische Wilhelms-University Münster, 48149 Münster, Germany
| | - M D Lange
- Institute of Physiology I, Westfälische Wilhelms-University Münster, 48149 Münster, Germany.
| |
Collapse
|
47
|
Meyer HC, Sangha S, Radley JJ, LaLumiere RT, Baratta MV. Environmental certainty influences the neural systems regulating responses to threat and stress. Neurosci Biobehav Rev 2021; 131:1037-1055. [PMID: 34673111 PMCID: PMC8642312 DOI: 10.1016/j.neubiorev.2021.10.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 10/20/2022]
Abstract
Flexible calibration of threat responding in accordance with the environment is an adaptive process that allows an animal to avoid harm while also maintaining engagement of other goal-directed actions. This calibration process, referred to as threat response regulation, requires an animal to calculate the probability that a given encounter will result in a threat so they can respond accordingly. Here we review the neural correlates of two highly studied forms of threat response suppression: extinction and safety conditioning. We focus on how relative levels of certainty or uncertainty in the surrounding environment alter the acquisition and application of these processes. We also discuss evidence indicating altered threat response regulation following stress exposure, including enhanced fear conditioning, and disrupted extinction and safety conditioning. To conclude, we discuss research using an animal model of coping that examines the impact of stressor controllability on threat responding, highlighting the potential for previous experiences with control, or other forms of coping, to protect against the effects of future adversity.
Collapse
Affiliation(s)
- Heidi C Meyer
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, 02215, USA.
| | - Susan Sangha
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| | - Jason J Radley
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, 52242, USA.
| | - Ryan T LaLumiere
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, 52242, USA.
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, USA.
| |
Collapse
|
48
|
Saeger HN, Olson DE. Psychedelic-inspired approaches for treating neurodegenerative disorders. J Neurochem 2021; 162:109-127. [PMID: 34816433 DOI: 10.1111/jnc.15544] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Psychedelics are increasingly being recognized for their potential to treat a wide range of brain disorders including depression, post-traumatic stress disorder (PTSD), and substance use disorder. Their broad therapeutic potential might result from an ability to rescue cortical atrophy common to many neuropsychiatric and neurodegenerative diseases by impacting neurotrophic factor gene expression, activating neuronal growth and survival mechanisms, and modulating the immune system. While the therapeutic potential of psychedelics has not yet been extended to neurodegenerative disorders, we provide evidence suggesting that approaches based on psychedelic science might prove useful for treating these diseases. The primary target of psychedelics, the 5-HT2A receptor, plays key roles in cortical neuron health and is dysregulated in Alzheimer's disease. Moreover, evidence suggests that psychedelics and related compounds could prove useful for treating the behavioral and psychological symptoms of dementia (BPSD). While more research is needed to probe the effects of psychedelics in models of neurodegenerative diseases, the robust effects of these compounds on structural and functional neuroplasticity and inflammation clearly warrant further investigation.
Collapse
Affiliation(s)
- Hannah N Saeger
- Pharmacology and Toxicology Graduate Group, University of California, Davis, Davis, California, USA
| | - David E Olson
- Department of Chemistry, University of California, Davis, Davis, California, USA.,Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA.,Center for Neuroscience, University of California, Davis, Davis, California, USA
| |
Collapse
|
49
|
Chakraborty P, Chattarji S, Jeanneteau F. A salience hypothesis of stress in PTSD. Eur J Neurosci 2021; 54:8029-8051. [PMID: 34766390 DOI: 10.1111/ejn.15526] [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: 07/10/2021] [Revised: 09/13/2021] [Accepted: 10/30/2021] [Indexed: 11/30/2022]
Abstract
Attention to key features of contexts and things is a necessary tool for all organisms. Detecting these salient features of cues, or simply, salience, can also be affected by exposure to traumatic stress, as has been widely reported in individuals suffering from post-traumatic stress disorder (PTSD). Interestingly, similar observations have been robustly replicated across many animal models of stress as well. By using evidence from such rodent stress paradigms, in the present review, we explore PTSD through the lens of salience processing. In this context, we propose that interaction between the neurotrophin brain-derived neurotrophic factor (BDNF) and glucocorticoids determines the long lasting cellular and behavioural consequences of stress salience. We also describe the dual effect of glucocorticoid therapy in the amelioration of PTSD symptoms. Finally, by integrating in vivo observations at multiple scales of plasticity, we propose a unifying hypothesis that pivots on a crucial role of glucocorticoid signalling in dynamically orchestrating stress salience.
Collapse
Affiliation(s)
- Prabahan Chakraborty
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France.,Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India
| | - Sumantra Chattarji
- Tata Institute of Fundamental Research, National Centre for Biological Sciences, Bellary Road, Bangalore, 560065, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Freddy Jeanneteau
- Institut de Genomique Fonctionnelle, University of Montpellier, Inserm, CNRS, Montpellier, 34090, France
| |
Collapse
|
50
|
Kefir ameliorates specific microbiota-gut-brain axis impairments in a mouse model relevant to autism spectrum disorder. Brain Behav Immun 2021; 97:119-134. [PMID: 34252569 DOI: 10.1016/j.bbi.2021.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/17/2021] [Accepted: 07/03/2021] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is one of the most severe developmental disorders, affecting on average 1 in 150 children worldwide. There is a great need for more effective strategies to improve quality of life in ASD subjects. The gut microbiome has emerged as a potential therapeutic target in ASD. A novel modulator of the gut microbiome, the traditionally fermented milk drink kefir, has recently been shown to modulate the microbiota and decrease repetitive behaviour, one of the hallmarks of ASD, in mice. As such, we hypothesized that kefir could ameliorate behavioural deficits in a mouse model relevant to ASD; the BTBR T+ Itpr3tf/J mouse strain. To this end, adult mice were administered either kefir (UK4) or a milk control for three weeks as treatment lead-in, after which they were assessed for their behavioural phenotype using a battery of tests. In addition, we assessed systemic immunity by flow cytometry and the gut microbiome using shotgun metagenomic sequencing. We found that indeed kefir decreased repetitive behaviour in this mouse model. Furthermore, kefir prolonged stress-induced increases in corticosterone 60 min post-stress, which was accompanied by an ameliorated innate immune response as measured by LY6Chi monocyte levels. In addition, kefir increased the levels of anti-inflammatory Treg cells in mesenteric lymph nodes (MLNs). Kefir also increased the relative abundance of Lachnospiraceae bacterium A2, which correlated with reduced repetitive behaviour and increased Treg cells in MLNs. Functionally, kefir modulated various predicted gut microbial pathways, including the gut-brain module S-Adenosylmethionine (SAM) synthesis, as well as L-valine biosynthesis and pyruvate fermentation to isobutanol, which all correlated with repetitive behaviour. Taken together our data show that kefir modulates peripheral immunoregulation, can ameliorate specific ASD behavioural dysfunctions and modulates selective aspects of the composition and function of the gut microbiome, indicating that kefir supplementation might prove a viable strategy in improving quality of life in ASD subjects.
Collapse
|