1
|
Jiang T, Feng M, Hutsell A, Lüscher B. Sex-specific GABAergic microcircuits that switch vulnerability into resilience to stress and reverse the effects of chronic stress exposure. Mol Psychiatry 2025; 30:2297-2308. [PMID: 39550416 PMCID: PMC12092295 DOI: 10.1038/s41380-024-02835-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024]
Abstract
Clinical and preclinical studies have identified somatostatin (SST)-positive interneurons as critical elements that regulate the vulnerability to stress-related psychiatric disorders. Conversely, disinhibition of SST neurons in mice results in resilience to the behavioral effects of chronic stress. Here, we established a low-dose chronic chemogenetic protocol to map these changes in positively and negatively motivated behaviors to specific brain regions. AAV-hM3Dq-mediated chronic activation of SST neurons in the prelimbic cortex (PLC) had antidepressant drug-like effects on anxiety- and anhedonia-like motivated behaviors in male but not female mice. Analogous manipulation of the ventral hippocampus (vHPC) had such effects in female but not male mice. Moreover, the activation of SST neurons in the PLC of male mice and the vHPC of female mice resulted in stress resilience. Activation of SST neurons in the PLC reversed prior chronic stress-induced defects in motivated behavior in males but was ineffective in females. Conversely, activation of SST neurons in the vHPC reversed chronic stress-induced behavioral alterations in females but not males. Quantitation of c-Fos+ and FosB+ neurons in chronic stress-exposed mice revealed that chronic activation of SST neurons leads to a paradoxical increase in pyramidal cell activity. Collectively, these data demonstrate that GABAergic microcircuits driven by dendrite targeting interneurons enable sex- and brain-region-specific neural plasticity that promotes stress resilience and reverses stress-induced anxiety- and anhedonia-like motivated behavior. The data provide a rationale for the lack of antidepressant efficacy of benzodiazepines and superior efficacy of dendrite-targeting, low-potency GABAA receptor agonists, independent of sex and despite striking sex differences in the relevant brain substrates.
Collapse
Affiliation(s)
- Tong Jiang
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Mengyang Feng
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexander Hutsell
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Bernhard Lüscher
- Department of Biology, Pennsylvania State University, University Park, PA, USA.
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA.
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
2
|
Liu S, Crawford J, Maltezos H, Sun Y, Tao R, Tao F. A glutamatergic brain neural circuit is critical for modulating trigeminal neuropathic pain. Pain 2025:00006396-990000000-00890. [PMID: 40310866 DOI: 10.1097/j.pain.0000000000003647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025]
Abstract
ABSTRACT Trigeminal neuropathic pain is a predominant symptom in patients with trigeminal neuralgia. However, the underlying neural circuit mechanism is still elusive. In this study, we investigated the role of a brain neural circuit in the modulation of trigeminal neuropathic pain. We used "Targeted Recombination in Active Populations" to identify activated neurons in brain structures. Anterograde and retrograde viral tracing combined with immunofluorescence staining was used to validate the activated neurons-involved neuronal pathway. We performed optogenetic stimulation and behavioral observation to dissect the brain neural circuitry that underlies the modulation of trigeminal neuropathic pain. We further conducted dual-color fiber photometry to analyze dynamic neurotransmitter release and real-time neuronal activity while observing pain behaviors simultaneously. We observed that mouse neurons in the anterior paraventricular nucleus of thalamus were activated specifically by chronic constriction injury of the infraorbital nerve. We further observed that specifical excitation or silencing of the activated neurons bidirectionally modulated the nerve injury-caused trigeminal neuropathic pain in mice. More importantly, optogenetic activation of the brain neural circuit from anterior paraventricular nucleus of thalamus to anterior cingulate cortex exacerbated such pain and this effect was blocked by an N-methyl-d-aspartate receptor antagonist. Meanwhile, optogenetic activation of this neural circuit markedly increased glutamate release and enhanced neuronal activity in the anterior cingulate cortex. Our results suggest that the identified brain neural circuit could be targeted to develop a novel neuromodulation therapy for trigeminal neuropathic pain.
Collapse
Affiliation(s)
- Sufang Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Joshua Crawford
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Hui Maltezos
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Yanjun Sun
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Ran Tao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United States
| |
Collapse
|
3
|
Dobolyi A. Integrating the COM-B model into behavioral neuroscience: A framework for understanding animal behavior. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111346. [PMID: 40154911 DOI: 10.1016/j.pnpbp.2025.111346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
In light of the intricate nature of animal behavior regulation, a theoretical model is proposed, grounded in the COM-B (Capability, Opportunity, Motivation - Behavior) framework, which has gained considerable traction in the domain of human behavioral intervention. When extending the COM-B model to behavioral neuroscience, we first discuss the utility of the model in animal research, particularly its capacity to integrate environmental and social factors, and enhance cross-species comparisons, including animal-to-human translations and evolutionary considerations. The subsequent discussion then summarizes the advantages of utilizing the COM-B model in neuroscience are summarized, including the facilitation of a systems-level understanding of behavior and the establishment of a link between neural mechanisms and specific behavioral components. The experimental design for the application of the COM-B model in neuroscience is proposed to elucidate the brain regulatory processes that govern behavior. Finally, three specific examples are provided to illustrate the theoretical considerations, namely feeding and social behavior, and the role of neuromodulators in the control of behavior.
Collapse
Affiliation(s)
- Arpád Dobolyi
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary; Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
4
|
Hsu LM, Shih YYI. Neuromodulation in Small Animal fMRI. J Magn Reson Imaging 2025; 61:1597-1617. [PMID: 39279265 PMCID: PMC11903207 DOI: 10.1002/jmri.29575] [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: 04/09/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/18/2024] Open
Abstract
The integration of functional magnetic resonance imaging (fMRI) with advanced neuroscience technologies in experimental small animal models offers a unique path to interrogate the causal relationships between regional brain activity and brain-wide network measures-a goal challenging to accomplish in human subjects. This review traces the historical development of the neuromodulation techniques commonly used in rodents, such as electrical deep brain stimulation, optogenetics, and chemogenetics, and focuses on their application with fMRI. We discuss their advantageousness roles in uncovering the signaling architecture within the brain and the methodological considerations necessary when conducting these experiments. By presenting several rodent-based case studies, we aim to demonstrate the potential of the multimodal neuromodulation approach in shedding light on neurovascular coupling, the neural basis of brain network functions, and their connections to behaviors. Key findings highlight the cell-type and circuit-specific modulation of brain-wide activity patterns and their behavioral correlates. We also discuss several future directions and feature the use of mediation and moderation analytical models beyond the intuitive evoked response mapping, to better leverage the rich information available in fMRI data with neuromodulation. Using fMRI alongside neuromodulation techniques provide insights into the mesoscopic (relating to the intermediate scale between single neurons and large-scale brain networks) and macroscopic fMRI measures that correlate with specific neuronal events. This integration bridges the gap between different scales of neuroscience research, facilitating the exploration and testing of novel therapeutic strategies aimed at altering network-mediated behaviors. In conclusion, the combination of fMRI with neuromodulation techniques provides crucial insights into mesoscopic and macroscopic brain dynamics, advancing our understanding of brain function in health and disease. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Li-Ming Hsu
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill
- Departments of Radiology, The University of North Carolina at Chapel Hill
| | - Yen-Yu Ian Shih
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill
- Departments of Neurology, The University of North Carolina at Chapel Hill
| |
Collapse
|
5
|
Hu B, Yin MY, Zhang CY, Shi Z, Wang L, Lei X, Li M, Li SW, Tuo QH. The INO80E at 16p11.2 locus increases risk of schizophrenia in humans and induces schizophrenia-like phenotypes in mice. EBioMedicine 2025; 114:105645. [PMID: 40088626 PMCID: PMC11957503 DOI: 10.1016/j.ebiom.2025.105645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Chromosome 16p11.2 is one of the most significant loci in the genome-wide association studies (GWAS) of schizophrenia. Despite several integrative analyses and functional genomics studies having been carried out to identify possible risk genes, their impacts in the pathogenesis of schizophrenia remain to be fully characterized. METHODS We performed expression quantitative trait loci (eQTL) and summary-data-based Mendelian randomization (SMR) analyses to identify schizophrenia risk genes in the 16p11.2 GWAS locus. We constructed a murine model with dysregulated expression of risk gene in the medial prefrontal cortex (mPFC) using stereotaxic injection of adeno-associated virus (AAV), followed by behavioural assessments, dendritic spine analyses and RNA sequencing. FINDINGS We identified significant associations between elevated INO80E mRNA expression in the frontal cortex and risk of schizophrenia. The mice overexpressing Ino80e in mPFC (Ino80e-OE) exhibited schizophrenia-like behaviours, including increased anxiety behaviour, anhedonia, and impaired prepulse inhibition (PPI) when compared with control group. The neuronal sparse labelling assay showed that the density of stubby spines in the pyramidal neurons of mPFC was significantly increased in Ino80e-OE mice compared with control mice. Transcriptomic analysis in the mPFC revealed significant alterations in the mRNA levels of schizophrenia-related genes and processes related to synapses upon overexpressing Ino80e. INTERPRETATION Our results suggest that upregulation of the Ino80e gene in mPFC may induce schizophrenia-like behaviours in mice, further supporting the hypothesis that INO80E is an authentic risk gene. FUNDING This project received support from the National Key Research and Development Program of China, National Natural Science Foundation of China, Key Research and Development Projects of Hunan Provincial Science and Technology Department, Science and Technology Innovation team of Hunan Province, etc.
Collapse
Affiliation(s)
- Bo Hu
- Hunan Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Mei-Yu Yin
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhe Shi
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Pharmacy of School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lu Wang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaoming Lei
- Hunan Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ming Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shi-Wu Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Qin-Hui Tuo
- Hunan Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
6
|
Coley AA, Batra K, Delahanty JM, Keyes LR, Pamintuan R, Ramot A, Hagemann J, Lee CR, Liu V, Adivikolanu H, Cressy J, Jia C, Massa F, LeDuke D, Gabir M, Durubeh B, Linderhof L, Patel R, Wichmann R, Li H, Fischer KB, Pereira T, Tye KM. Predicting Future Development of Stress-Induced Anhedonia From Cortical Dynamics and Facial Expression. RESEARCH SQUARE 2025:rs.3.rs-5537951. [PMID: 40166035 PMCID: PMC11957211 DOI: 10.21203/rs.3.rs-5537951/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The current state of mental health treatment for individuals diagnosed with major depressive disorder leaves billions of individuals with first-line therapies that are ineffective or burdened with undesirable side effects. One major obstacle is that distinct pathologies may currently be diagnosed as the same disease and prescribed the same treatments. The key to developing antidepressants with ubiquitous efficacy is to first identify a strategy to differentiate between heterogeneous conditions. Major depression is characterized by hallmark features such as anhedonia and a loss of motivation1,2, and it has been recognized that even among inbred mice raised under identical housing conditions, we observe heterogeneity in their susceptibility and resilience to stress3. Anhedonia, a condition identified in multiple neuropsychiatric disorders, is described as the inability to experience pleasure and is linked to anomalous medial prefrontal cortex (mPFC) activity4. The mPFC is responsible for higher order functions5-8, such as valence encoding; however, it remains unknown how mPFC valence-specific neuronal population activity is affected during anhedonic conditions. To test this, we implemented the unpredictable chronic mild stress (CMS) protocol9-11 in mice and examined hedonic behaviors following stress and ketamine treatment. We used unsupervised clustering to delineate individual variability in hedonic behavior in response to stress. We then performed in vivo 2-photon calcium imaging to longitudinally track mPFC valence-specific neuronal population dynamics during a Pavlovian discrimination task. Chronic mild stress mice exhibited a blunted effect in the ratio of mPFC neural population responses to rewards relative to punishments after stress that rebounds following ketamine treatment. Also, a linear classifier revealed that we can decode susceptibility to chronic mild stress based on mPFC valence-encoding properties prior to stress-exposure and behavioral expression of susceptibility. Lastly, we utilized markerless pose tracking computer vision tools to predict whether a mouse would become resilient or susceptible based on facial expressions during a Pavlovian discrimination task. These results indicate that mPFC valence encoding properties and behavior are predictive of anhedonic states. Altogether, these experiments point to the need for increased granularity in the measurement of both behavior and neural activity, as these factors can predict the predisposition to stress-induced anhedonia.
Collapse
Affiliation(s)
- Austin A. Coley
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, Los Angeles, Los Angeles, CA
| | - Kanha Batra
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Jeremy M. Delahanty
- The Salk Institute for Biological Studies, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
| | - Laurel R. Keyes
- The Salk Institute for Biological Studies, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
| | - Rachelle Pamintuan
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Assaf Ramot
- University of California, San Diego, La Jolla, CA
| | | | - Christopher R. Lee
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Neuroscience Graduate Program, University of California, San Diego, La Jolla
- Howard Hughes Medical Institute, La Jolla, CA
| | - Vivian Liu
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Harini Adivikolanu
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Jianna Cressy
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Medical Scientist Training Program, University of California, San Diego, La Jolla
| | - Caroline Jia
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Medical Scientist Training Program, University of California, San Diego, La Jolla
| | - Francesca Massa
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Deryn LeDuke
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Moumen Gabir
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Bra’a Durubeh
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Lexe Linderhof
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Reesha Patel
- The Salk Institute for Biological Studies, La Jolla, CA
- Northwestern University, Chicago, IL
| | - Romy Wichmann
- The Salk Institute for Biological Studies, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
| | - Hao Li
- The Salk Institute for Biological Studies, La Jolla, CA
- Northwestern University, Chicago, IL
| | | | - Talmo Pereira
- The Salk Institute for Biological Studies, La Jolla, CA
| | - Kay M. Tye
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
- Kavli Institute for the Brain and Mind
| |
Collapse
|
7
|
Pickup E, Weber F. Sleep circuits welcome the cortex. Sleep 2025; 48:zsae312. [PMID: 39756401 PMCID: PMC11893530 DOI: 10.1093/sleep/zsae312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Indexed: 01/07/2025] Open
Affiliation(s)
- Emily Pickup
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Franz Weber
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
Varela RB, Macpherson H, Walker AJ, Houghton T, Yates C, Yates NJ, Daygon VD, Tye SJ. Inflammation and metabolic dysfunction underly anhedonia-like behavior in antidepressant resistant male rats. Brain Behav Immun 2025; 127:170-182. [PMID: 40064431 DOI: 10.1016/j.bbi.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 02/17/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Inflammation and metabolic dysfunction impair dopamine neurotransmission, which is thought to serve as a critical mechanism underpinning motivational deficits such as anhedonia across a range of psychiatric and neurological disorders. This difficult-to-treat transdiagnostic symptom has important implications for treatment resistant depression (TRD), and may warrant more targeted therapeutic approaches that address the underlying pathophysiological mechanisms. Using the adrenocorticotrophic hormone (ACTH) model of antidepressant treatment resistance we characterized the relationship between antidepressant-like and anhedonia-like behavioral responses to bupropion, mesocortical tyrosine hydroxylase (TH) expression, chronic low-grade inflammation, and metabolic changes in male rats. We demonstrate that chronic ACTH elicited both an antidepressant resistant- and anhedonia-like phenotype in forced swim and effort-related choice behavioral tasks, respectively. This was associated with decreased TH expression in the brain, increased central and peripheral markers of inflammation, and peripheral metabolic disturbances, including impairment of immune cell insulin action. Multivariate analysis revealed that peripheral interleukin-6 (IL-6) levels, immune cell glucose uptake and disturbance of nucleotide metabolism were strongly associated with anhedonia-like behavior. Post-hoc analyses further confirmed strong correlations between TH expression, inflammation and behavioral performance. These data suggest that stress hormone-induced upregulation of inflammation concurrent with the impairment of insulin-mediated glucose uptake into immune cells is associated with disruption of nucleotide metabolism, and potential impaired central dopamine synthesis contributing to the behavioral expression of anhedonia. These results suggest that immunometabolic perturbations concomitant with impaired insulin action at the level of the immune cell result in a metabolically deficient state that directly impacts nucleotide precursors essential for dopamine synthesis and effortful behavior. These results highlight the potential for immune and metabolic markers for individualized treatment of refractory depression and anhedonia.
Collapse
Affiliation(s)
- Roger B Varela
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.
| | - Heather Macpherson
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Adam J Walker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States; Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Tristan Houghton
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia; Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Clarissa Yates
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Nathanael J Yates
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Venea D Daygon
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Susannah J Tye
- Functional Neuromodulation and Novel Therapeutics Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States; Department of Psychiatry and Behavioral Science, Emory University, Atlanta, GA, United States.
| |
Collapse
|
9
|
Zhou XA, Jiang Y, Gomez-Cid L, Yu X. Elucidating hemodynamics and neuro-glio-vascular signaling using rodent fMRI. Trends Neurosci 2025; 48:227-241. [PMID: 39843335 PMCID: PMC11903151 DOI: 10.1016/j.tins.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/09/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025]
Abstract
Despite extensive functional mapping studies using rodent functional magnetic resonance imaging (fMRI), interpreting the fMRI signals in relation to their neuronal origins remains challenging due to the hemodynamic nature of the response. Ultra high-resolution rodent fMRI, beyond merely enhancing spatial specificity, has revealed vessel-specific hemodynamic responses, highlighting the distinct contributions of intracortical arterioles and venules to fMRI signals. This 'single-vessel' fMRI approach shifts the paradigm of rodent fMRI, enabling its integration with other neuroimaging modalities to investigate neuro-glio-vascular (NGV) signaling underlying a variety of brain dynamics. Here, we review the emerging trend of combining multimodal fMRI with opto/chemogenetic neuromodulation and genetically encoded biosensors for cellular and circuit-specific recording, offering unprecedented opportunities for cross-scale brain dynamic mapping in rodent models.
Collapse
Affiliation(s)
- Xiaoqing Alice Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | - Yuanyuan Jiang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Lidia Gomez-Cid
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Xin Yu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
10
|
Azarfarin M, Shahla MM, Mohaddes G, Dadkhah M. Non-pharmacological therapeutic paradigms in stress-induced depression: from novel therapeutic perspective with focus on cell-based strategies. Acta Neuropsychiatr 2025; 37:e10. [PMID: 39973753 DOI: 10.1017/neu.2024.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Major depressive disorder (MDD) is considered a psychiatric disorder and have a relationship with stressful events. Although the common therapeutic approaches against MDD are diverse, a large number of patients do not present an adequate response to antidepressant treatments. On the other hand, effective non-pharmacological treatments for MDD and their tolerability are addressed. Several affective treatments for MDD are used but non-pharmacological strategies for decreasing the common depression-related drugs side effects have been focused recently. However, the potential of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs), microRNAs (miRNAs) as cell-based therapeutic paradigms, besides other non-pharmacological strategies including mitochondrial transfer, plasma, transcranial direct current stimulation (tDCS), transcranial magnetic stimulation (TMS), and exercise therapy needs to further study. This review explores the therapeutic potential of cell-based therapeutic non-pharmacological paradigms for MDD treatment. In addition, plasma therapy, mitotherapy, and exercise therapy in several in vitro and in vivo conditions in experimental disease models along with tDCS and TMS will be discussed as novel non-pharmacological promising therapeutic approaches.
Collapse
Affiliation(s)
- Maryam Azarfarin
- Neuroscience Research center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Neuroscience, Faculty of Advanced Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Gisou Mohaddes
- Neuroscience Research center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biomedical Education, College of Osteopathic Medicine, California Health Sciences University, Clovis, CA, USA
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Neuroscience Research Group, Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
11
|
Hike D, Liu X, Xie Z, Zhang B, Choi S, Zhou XA, Liu A, Murstein A, Jiang Y, Devor A, Yu X. High-resolution awake mouse fMRI at 14 tesla. eLife 2025; 13:RP95528. [PMID: 39786364 PMCID: PMC11717365 DOI: 10.7554/elife.95528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
High-resolution awake mouse functional magnetic resonance imaging (fMRI) remains challenging despite extensive efforts to address motion-induced artifacts and stress. This study introduces an implantable radio frequency (RF) surface coil design that minimizes image distortion caused by the air/tissue interface of mouse brains while simultaneously serving as a headpost for fixation during scanning. Furthermore, this study provides a thorough acclimation method used to accustom animals to the MRI environment minimizing motion-induced artifacts. Using a 14 T scanner, high-resolution fMRI enabled brain-wide functional mapping of visual and vibrissa stimulation at 100 µm×100 µm×200 µm resolution with a 2 s per frame sampling rate. Besides activated ascending visual and vibrissa pathways, robust blood oxygen level-dependent (BOLD) responses were detected in the anterior cingulate cortex upon visual stimulation and spread through the ventral retrosplenial area (VRA) with vibrissa air-puff stimulation, demonstrating higher-order sensory processing in association cortices of awake mice. In particular, the rapid hemodynamic responses in VRA upon vibrissa stimulation showed a strong correlation with the hippocampus, thalamus, and prefrontal cortical areas. Cross-correlation analysis with designated VRA responses revealed early positive BOLD signals at the contralateral barrel cortex (BC) occurring 2 s prior to the air-puff in awake mice with repetitive stimulation, which was not detected using a randomized stimulation paradigm. This early BC activation indicated a learned anticipation through the vibrissa system and association cortices in awake mice under continuous exposure of repetitive air-puff stimulation. This work establishes a high-resolution awake mouse fMRI platform, enabling brain-wide functional mapping of sensory signal processing in higher association cortical areas.
Collapse
Affiliation(s)
- David Hike
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| | - Xiaochen Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| | - Zeping Xie
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| | - Bei Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| | - Sangcheon Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| | - Xiaoqing Alice Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| | - Andy Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
- Graduate Program in Neuroscience, Boston UniversityBostonUnited States
| | - Alyssa Murstein
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
- Graduate Program in Neuroscience, Boston UniversityBostonUnited States
| | - Yuanyuan Jiang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| | - Anna Devor
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
- Department of Biomedical Engineering, Boston UniversityBostonUnited States
| | - Xin Yu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General HospitalCharlestownUnited States
| |
Collapse
|
12
|
Kovacevic N, Meghdadi A, Berka C. Characterizing PTSD Using Electrophysiology: Towards A Precision Medicine Approach. Clin EEG Neurosci 2025:15500594241309680. [PMID: 39763472 DOI: 10.1177/15500594241309680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Objective. Resting-state EEG measures have shown potential in distinguishing individuals with PTSD from healthy controls. ERP components such as N2, P3, and late positive potential have been consistently linked to cognitive abnormalities in PTSD, especially in tasks involving emotional or trauma-related stimuli. However, meta-analyses have reported inconsistent findings. The understanding of biomarkers that can classify the varied symptoms of PTSD remains limited. This study aimed to develop a concise set of electrophysiological biomarkers, using neutral cognitive tasks, that could be applied across psychiatric conditions, and to identify biomarkers associated with the anxiety and depression dimensions of PTSD. Approach. Continuous simultaneous recordings of EEG and electrocardiogram (ECG) were obtained in veterans with PTSD (n = 29) and healthy controls (n = 62) during computerized tasks. EEG, ERP, and heart rate measures were evaluated in terms of their ability to discriminate between the groups or correlate with psychological measures. Results. The PTSD cohort exhibited faster alpha oscillations, reduced alpha power, and a flatter power spectrum. Furthermore, stronger reduction in alpha power was associated with higher trait anxiety, while a flatter slope was related to more severe depression symptoms in individuals with PTSD. In ERP tasks of visual memory and sustained attention, the PTSD cohort demonstrated delayed and exaggerated early components, along with attenuated LPP amplitudes. The three tasks revealed distinct and complementary EEG signatures PTSD. Significance. Multimodal individualized biomarkers based on EEG, cognitive ERPs, and ECG show promise as objective tools for assessing mood and anxiety disturbances within the PTSD spectrum.
Collapse
Affiliation(s)
| | | | - Chris Berka
- Advanced Brain Monitoring, Carlsbad, CA, USA
| |
Collapse
|
13
|
Duarte JMN. Challenges of Investigating Compartmentalized Brain Energy Metabolism Using Nuclear Magnetic Resonance Spectroscopy in vivo. Neurochem Res 2025; 50:73. [PMID: 39754627 PMCID: PMC11700056 DOI: 10.1007/s11064-024-04324-4] [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: 08/05/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/06/2025]
Abstract
Brain function requires continuous energy supply. Thus, unraveling brain metabolic regulation is critical not only for our basic understanding of overall brain function, but also for the cellular basis of functional neuroimaging techniques. While it is known that brain energy metabolism is exquisitely compartmentalized between astrocytes and neurons, the metabolic and neuro-energetic basis of brain activity is far from fully understood. 1H nuclear magnetic resonance (NMR) spectroscopy has been widely used to detect variations in metabolite levels, including glutamate and GABA, while 13C NMR spectroscopy has been employed to study metabolic compartmentation and to determine metabolic rates coupled brain activity, focusing mainly on the component corresponding to excitatory glutamatergic neurotransmission. The rates of oxidative metabolism in neurons and astrocytes are both associated with the rate of the glutamate-glutamine cycle between neurons and astrocytes. However, any possible correlation between energy metabolism pathways and the inhibitory GABAergic neurotransmission rate in the living brain remains to be experimentally demonstrated. That is due to low GABA levels, and the consequent challenge of determining GABAergic rates in a non-invasive manner. This brief review surveys the state-of-the-art analyses of energy metabolism in neurons and astrocytes contributing to glutamate and GABA synthesis using 13C NMR spectroscopy in vivo, and identifies limitations that need to be overcome in future studies.
Collapse
Affiliation(s)
- João M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Sinclair LI, Ajram L, Seneviratne G, Tracy D, Critchley H. Why does research matter to psychiatrists? Brain Neurosci Adv 2025; 9:23982128241305866. [PMID: 39803635 PMCID: PMC11724414 DOI: 10.1177/23982128241305866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Affiliation(s)
- Lindsey I Sinclair
- Dementia Research Group, Bristol Medical School, University of Bristol, Bristol, UK
- Academic Faculty, Royal College of Psychiatrists, London, UK
| | - Laura Ajram
- British Neuroscience Association, Bristol, UK
| | - Gertrude Seneviratne
- South London and Maudsley NHS Foundation Trust, London, UK
- South London Partnership, London, UK
| | - Derek Tracy
- South London and Maudsley NHS Foundation Trust, London, UK
- Brunel University Medical School, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Department of Psychiatry, Imperial College London, London, UK
| | - Hugo Critchley
- Academic Faculty, Royal College of Psychiatrists, London, UK
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
15
|
Huang X, Xi C, Fang Y, Ye R, Wang X, Zhang S, Cui Y, Guo Y, Zhang J, Ji GJ, Zhu C, Luo Y, Chen X, Wang K, Tian Y, Yu F. Therapeutic Efficacy of Reward Circuit‐Targeted Transcranial Magnetic Stimulation (TMS) on Suicidal Ideation in Depressed Patients: A Sham‐Controlled Trial of Two TMS Protocols. Depress Anxiety 2025; 2025. [DOI: 10.1155/da/1767477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 12/13/2024] [Indexed: 05/04/2025] Open
Abstract
Background: Suicide is one of the leading causes of premature death, and dysfunctional reward processing may serve as a potential mechanism. However, effective treatment targeting reward circuits is rarely reported.Objective: The present study investigated the therapeutic efficacy of two individualized protocols, repetitive transcranial magnetic stimulation (rTMS) and intermittent theta burst stimulation (iTBS), targeting the left dorsolateral prefrontal cortex (lDLPFC)–nucleus accumbens (NAcc) circuit on suicidal ideation among patients with major depressive disorder (MDD).Methods: Here, 40 healthy controls (HCs) and 70 MDD patients (MDDs) were recruited for this double‐blinded, sham‐controlled clinical trial. The reward learning process during the Iowa gambling task (IGT) was initially measured at the baseline. Further, 62 MDDs were assigned to receive 15 daily sessions of individualized rTMS (n = 25), iTBS (n = 15), or sham treatment (n = 22) to the site of strongest lDLPFC–NAcc connectivity.Results: We found MDDs demonstrated abnormalities in both IGT performance and reward‐associated event‐related potential (ERP) components compared to HCs. MDDs in the rTMS and iTBS groups showed significant improvements in suicidal ideation and anhedonia symptoms compared to the sham group. The rTMS group also exhibited a more negative‐going N170 and feedback‐related negativity (FRN) after treatment, and the increase in N170 absolute amplitude posttreatment showed a trend of correlation with improved Temporal Experience Pleasure Scales (TEPSs) and TEPS‐anticipatory (TEPS‐ant) scores.Conclusion: The current study indicates that reward circuit‐based rTMS and iTBS showed comparable antisuicidal effects in depressive patients, suggesting that the lDLPFC–NAcc pathway may serve as a potential treatment target.Trial Registration: ClinicalTrials.gov identifier: NCT03991572
Collapse
|
16
|
Coley AA, Batra K, Delahanty JM, Keyes LR, Pamintuan R, Ramot A, Hagemann J, Lee CR, Liu V, Adivikolanu H, Cressy J, Jia C, Massa F, LeDuke D, Gabir M, Durubeh B, Linderhof L, Patel R, Wichmann R, Li H, Fischer KB, Pereira T, Tye KM. Predicting Future Development of Stress-Induced Anhedonia From Cortical Dynamics and Facial Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.629202. [PMID: 39764017 PMCID: PMC11702711 DOI: 10.1101/2024.12.18.629202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The current state of mental health treatment for individuals diagnosed with major depressive disorder leaves billions of individuals with first-line therapies that are ineffective or burdened with undesirable side effects. One major obstacle is that distinct pathologies may currently be diagnosed as the same disease and prescribed the same treatments. The key to developing antidepressants with ubiquitous efficacy is to first identify a strategy to differentiate between heterogeneous conditions. Major depression is characterized by hallmark features such as anhedonia and a loss of motivation1,2, and it has been recognized that even among inbred mice raised under identical housing conditions, we observe heterogeneity in their susceptibility and resilience to stress3. Anhedonia, a condition identified in multiple neuropsychiatric disorders, is described as the inability to experience pleasure and is linked to anomalous medial prefrontal cortex (mPFC) activity4. The mPFC is responsible for higher order functions5-8, such as valence encoding; however, it remains unknown how mPFC valence-specific neuronal population activity is affected during anhedonic conditions. To test this, we implemented the unpredictable chronic mild stress (CMS) protocol9-11 in mice and examined hedonic behaviors following stress and ketamine treatment. We used unsupervised clustering to delineate individual variability in hedonic behavior in response to stress. We then performed in vivo 2-photon calcium imaging to longitudinally track mPFC valence-specific neuronal population dynamics during a Pavlovian discrimination task. Chronic mild stress mice exhibited a blunted effect in the ratio of mPFC neural population responses to rewards relative to punishments after stress that rebounds following ketamine treatment. Also, a linear classifier revealed that we can decode susceptibility to chronic mild stress based on mPFC valence-encoding properties prior to stress-exposure and behavioral expression of susceptibility. Lastly, we utilized markerless pose tracking computer vision tools to predict whether a mouse would become resilient or susceptible based on facial expressions during a Pavlovian discrimination task. These results indicate that mPFC valence encoding properties and behavior are predictive of anhedonic states. Altogether, these experiments point to the need for increased granularity in the measurement of both behavior and neural activity, as these factors can predict the predisposition to stress-induced anhedonia.
Collapse
Affiliation(s)
- Austin A. Coley
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, Los Angeles, Los Angeles, CA
| | - Kanha Batra
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Jeremy M. Delahanty
- The Salk Institute for Biological Studies, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
| | - Laurel R. Keyes
- The Salk Institute for Biological Studies, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
| | - Rachelle Pamintuan
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Assaf Ramot
- University of California, San Diego, La Jolla, CA
| | | | - Christopher R. Lee
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Neuroscience Graduate Program, University of California, San Diego, La Jolla
- Howard Hughes Medical Institute, La Jolla, CA
| | - Vivian Liu
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Harini Adivikolanu
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Jianna Cressy
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Medical Scientist Training Program, University of California, San Diego, La Jolla
| | - Caroline Jia
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Medical Scientist Training Program, University of California, San Diego, La Jolla
| | - Francesca Massa
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Deryn LeDuke
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Moumen Gabir
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Bra’a Durubeh
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Lexe Linderhof
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
| | - Reesha Patel
- The Salk Institute for Biological Studies, La Jolla, CA
- Northwestern University, Chicago, IL
| | - Romy Wichmann
- The Salk Institute for Biological Studies, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
| | - Hao Li
- The Salk Institute for Biological Studies, La Jolla, CA
- Northwestern University, Chicago, IL
| | | | - Talmo Pereira
- The Salk Institute for Biological Studies, La Jolla, CA
| | - Kay M. Tye
- The Salk Institute for Biological Studies, La Jolla, CA
- University of California, San Diego, La Jolla, CA
- Howard Hughes Medical Institute, La Jolla, CA
- Kavli Institute for the Brain and Mind
| |
Collapse
|
17
|
Cattarinussi G, Heidari-Foroozan M, Jafary H, Mohammadi E, Sambataro F, Ferro A, Barone Y, Delvecchio G. Resting-state functional magnetic resonance imaging alterations in first-degree relatives of individuals with bipolar disorder: A systematic review. J Affect Disord 2024; 365:321-331. [PMID: 39142577 DOI: 10.1016/j.jad.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Relatives of individuals with bipolar disorder (BD) are at higher risk of developing the disorder. Identifying brain alterations associated with familial vulnerability in BD can help discover endophenotypes, which are quantifiable biological traits more prevalent in unaffected relatives of BD (BD-RELs) than the general population. This review aimed at expanding our knowledge on endophenotypes of BD by providing an overview of resting-state functional magnetic resonance imaging (rs-fMRI) alterations in BD-RELs. METHODS A systematic search of PubMed, Scopus, and Web of Science was performed to identify all available rs-fMRI studies conducted in BD-RELs up to January 2024. A total of 18 studies were selected. Six included BD-RELs with no history of psychiatric disorders and 10 included BD-RELs that presented psychiatric disorders. Two investigations examined rs-fMRI alterations in BD-RELs with and without subthreshold symptoms for BD. RESULTS BD-RELs presented rs-fMRI alterations in the cortico-limbic network, fronto-thalamic-striatal circuit, fronto-occipital network, and, to a lesser extent, in the default mode network. This was true both for BD-RELs with no history of psychopathology and for BD-RELs that presented psychiatric disorders. The direct comparison of rs-fMRI alterations in BD-RELs with and without psychiatric symptoms displayed largely non-overlapping patterns of rs-fMRI abnormalities. LIMITATIONS Small sample sizes and the clinical heterogeneity of BD-RELs limit the generalizability of our findings. CONCLUSIONS The current literature suggests that first-degree BD-RELs exhibit rs-fMRI alterations in brain circuits involved in emotion regulation, cognition, reward processing, and psychosis susceptibility. Future studies are needed to validate these findings and to explore their potential as biomarkers for early detection and intervention.
Collapse
Affiliation(s)
- Giulia Cattarinussi
- Department of Neuroscience (DNS), Padua Neuroscience Center, University of Padova, Padua, Italy; Padua Neuroscience Center, University of Padova, Padua, Italy
| | - Mahsa Heidari-Foroozan
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hosein Jafary
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Esmaeil Mohammadi
- Department of Neurological Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Fabio Sambataro
- Department of Neuroscience (DNS), Padua Neuroscience Center, University of Padova, Padua, Italy; Padua Neuroscience Center, University of Padova, Padua, Italy
| | - Adele Ferro
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ylenia Barone
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Giuseppe Delvecchio
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
18
|
Hu YB, Deng X, Liu L, Cao CC, Su YW, Gao ZJ, Cheng X, Kong D, Li Q, Shi YW, Wang XG, Ye X, Zhao H. Distinct roles of excitatory and inhibitory neurons in the medial prefrontal cortex in the expression and reconsolidation of methamphetamine-associated memory in male mice. Neuropsychopharmacology 2024; 49:1827-1838. [PMID: 38730034 PMCID: PMC11473735 DOI: 10.1038/s41386-024-01879-2] [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: 12/06/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024]
Abstract
Methamphetamine, a commonly abused drug, is known for its high relapse rate. The persistence of addictive memories associated with methamphetamine poses a significant challenge in preventing relapse. Memory retrieval and subsequent reconsolidation provide an opportunity to disrupt addictive memories. However, the key node in the brain network involved in methamphetamine-associated memory retrieval has not been clearly defined. In this study, using the conditioned place preference in male mice, whole brain c-FOS mapping and functional connectivity analysis, together with chemogenetic manipulations of neural circuits, we identified the medial prefrontal cortex (mPFC) as a critical hub that integrates inputs from the retrosplenial cortex and the ventral tegmental area to support both the expression and reconsolidation of methamphetamine-associated memory during its retrieval. Surprisingly, with further cell-type specific analysis and manipulation, we also observed that methamphetamine-associated memory retrieval activated inhibitory neurons in the mPFC to facilitate memory reconsolidation, while suppressing excitatory neurons to aid memory expression. These findings provide novel insights into the neural circuits and cellular mechanisms involved in the retrieval process of addictive memories. They suggest that targeting the balance between excitation and inhibition in the mPFC during memory retrieval could be a promising treatment strategy to prevent relapse in methamphetamine addiction.
Collapse
Affiliation(s)
- Yu-Bo Hu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xi Deng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lu Liu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Can-Can Cao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ya-Wen Su
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhen-Jie Gao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Deshan Kong
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Qi Li
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan-Wei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiao-Guang Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Hu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
19
|
Qiao Y, Lu H, Yang Y, Zang Y. Neuronal basis of high frequency fMRI fluctuation: direct evidence from simultaneous recording. Front Hum Neurosci 2024; 18:1501310. [PMID: 39545149 PMCID: PMC11560898 DOI: 10.3389/fnhum.2024.1501310] [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: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 11/17/2024] Open
Abstract
Resting-state functional magnetic resonance imaging (RS-fMRI) has been extensively utilized for noninvasive investigation of human brain activity. While studies employing simultaneous recordings of fMRI and electrophysiology have established a connection between the low-frequency fluctuation (< 0.1 Hz) observed in RS-fMRI and the local field potential (LFP), it remains unclear whether the RS-fMRI signal exhibits frequency-dependent modulation, which is a well-documented phenomenon in LFP. The present study concurrently recorded resting-state functional magnetic resonance imaging (RS-fMRI) and local field potentials (LFP) in the striatum of 8 rats before and after a pharmacological manipulation. We observed a highly similar frequency-dependent pattern of amplitude changes in both RS-fMRI and LFP following the manipulation, specifically an increase in high-frequency band amplitudes accompanied by a decrease in low-frequency band amplitudes. These findings provide direct evidence that the enhanced high-frequency fluctuations and reduced low-frequency fluctuations observed in RS-fMRI may reflect heightened neuronal activity.
Collapse
Affiliation(s)
- Yang Qiao
- Centre for Cognitive and Brain Sciences, University of Macau, Macao SAR, China
- Faculty of Health Sciences, University of Macau, Macao SAR, China
- Centre for Cognition and Brain Disorders/Department of Neurology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
- Institute of Psychological Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, Zhejiang, China
| | - Hanbing Lu
- Neuroimaging Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MA, United States
| | - Yihong Yang
- Neuroimaging Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MA, United States
| | - Yufeng Zang
- Centre for Cognition and Brain Disorders/Department of Neurology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
- Institute of Psychological Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, Zhejiang, China
- TMS Center, Deqing Hospital of Hangzhou Normal University, Huzhou, Zhejiang, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric disease, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
20
|
Long C, Lee K, Yang L, Dafalias T, Wu AK, Masmanidis SC. Constraints on the subsecond modulation of striatal dynamics by physiological dopamine signaling. Nat Neurosci 2024; 27:1977-1986. [PMID: 38961230 PMCID: PMC11608082 DOI: 10.1038/s41593-024-01699-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
Dopaminergic neurons play a crucial role in associative learning, but their capacity to regulate behavior on subsecond timescales remains debated. It is thought that dopaminergic neurons drive certain behaviors by rapidly modulating striatal spiking activity; however, a view has emerged that only artificially high (that is, supra-physiological) dopamine signals alter behavior on fast timescales. This raises the possibility that moment-to-moment striatal spiking activity is not strongly shaped by dopamine signals in the physiological range. To test this, we transiently altered dopamine levels while monitoring spiking responses in the ventral striatum of behaving mice. These manipulations led to only weak changes in striatal activity, except when dopamine release exceeded reward-matched levels. These findings suggest that dopaminergic neurons normally play a minor role in the subsecond modulation of striatal dynamics in relation to other inputs and demonstrate the importance of discerning dopaminergic neuron contributions to brain function under physiological and potentially nonphysiological conditions.
Collapse
Affiliation(s)
- Charltien Long
- Department of Neurobiology, University of California, Los Angeles, CA, USA
- Medical Scientist Training Program, University of California, Los Angeles, CA, USA
| | - Kwang Lee
- Department of Neurobiology, University of California, Los Angeles, CA, USA
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Long Yang
- Department of Neurobiology, University of California, Los Angeles, CA, USA
| | - Theresia Dafalias
- Department of Neurobiology, University of California, Los Angeles, CA, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Alexander K Wu
- Department of Neurobiology, University of California, Los Angeles, CA, USA
| | - Sotiris C Masmanidis
- Department of Neurobiology, University of California, Los Angeles, CA, USA.
- California Nanosystems Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
21
|
Hike D, Liu X, Xie Z, Zhang B, Choi S, Zhou XA, Liu A, Murstein A, Jiang Y, Devor A, Yu X. High-resolution awake mouse fMRI at 14 Tesla. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.08.570803. [PMID: 38106227 PMCID: PMC10723470 DOI: 10.1101/2023.12.08.570803] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
High-resolution awake mouse fMRI remains challenging despite extensive efforts to address motion-induced artifacts and stress. This study introduces an implantable radiofrequency (RF) surface coil design that minimizes image distortion caused by the air/tissue interface of mouse brains while simultaneously serving as a headpost for fixation during scanning. Furthermore, this study provides a thorough acclimation method used to accustom animals to the MRI environment minimizing motion induced artifacts. Using a 14T scanner, high-resolution fMRI enabled brain-wide functional mapping of visual and vibrissa stimulation at 100×100×200μm resolution with a 2s per frame sampling rate. Besides activated ascending visual and vibrissa pathways, robust BOLD responses were detected in the anterior cingulate cortex upon visual stimulation and spread through the ventral retrosplenial area (VRA) with vibrissa air-puff stimulation, demonstrating higher-order sensory processing in association cortices of awake mice. In particular, the rapid hemodynamic responses in VRA upon vibrissa stimulation showed a strong correlation with the hippocampus, thalamus, and prefrontal cortical areas. Cross-correlation analysis with designated VRA responses revealed early positive BOLD signals at the contralateral barrel cortex (BC) occurring 2 seconds prior to the air-puff in awake mice with repetitive stimulation, which was not detected using a randomized stimulation paradigm. This early BC activation indicated a learned anticipation through the vibrissa system and association cortices in awake mice under continuous training of repetitive air-puff stimulation. This work establishes a high-resolution awake mouse fMRI platform, enabling brain-wide functional mapping of sensory signal processing in higher association cortical areas.
Collapse
Affiliation(s)
- David Hike
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| | - Xiaochen Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| | - Zeping Xie
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| | - Bei Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| | - Sangcheon Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| | - Xiaoqing Alice Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| | - Andy Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
- Graduate program in Neuroscience, Boston University, 610 Commonwealth Avenue, Boston, Massachusetts, USA, 02215
| | - Alyssa Murstein
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
- Graduate program in Neuroscience, Boston University, 610 Commonwealth Avenue, Boston, Massachusetts, USA, 02215
| | - Yuanyuan Jiang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| | - Anna Devor
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
- Department of Biomedical Engineering, Boston University, 610 Commonwealth Avenue, Boston, Massachusetts, USA, 02215
| | - Xin Yu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 Thirteenth Street, Charlestown, Massachusetts, USA 02129
| |
Collapse
|
22
|
Isaac J, Karkare SC, Balasubramanian H, Schappaugh N, Javier JL, Rashid M, Murugan M. Sex differences in neural representations of social and nonsocial reward in the medial prefrontal cortex. Nat Commun 2024; 15:8018. [PMID: 39271723 PMCID: PMC11399386 DOI: 10.1038/s41467-024-52294-6] [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: 04/23/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
The reinforcing nature of social interactions is necessary for the maintenance of appropriate social behavior. However, the neural substrates underlying social reward processing and how they might differ based on the sex and internal state of the animal remains unknown. It is also unclear whether these neural substrates are shared with those involved in nonsocial rewarding processing. We developed a fully automated, two choice (social-sucrose) operant assay in which mice choose between social and nonsocial rewards to directly compare the reward-related behaviors associated with two competing stimuli. We performed cellular resolution calcium imaging of medial prefrontal cortex (mPFC) neurons in male and female mice across varying states of water restriction and social isolation. We found that mPFC neurons maintain largely non-overlapping, flexible representations of social and nonsocial reward that vary with internal state in a sex-dependent manner. Additionally, optogenetic manipulation of mPFC activity during the reward period of the assay disrupted reward-seeking behavior across male and female mice. Thus, using a two choice operant assay, we have identified sex-dependent, non-overlapping neural representations of social and nonsocial reward in the mPFC that vary with internal state and that are essential for appropriate reward-seeking behavior.
Collapse
Affiliation(s)
- Jennifer Isaac
- Neuroscience Graduate Program, Emory University, Atlanta, GA, 30322, USA
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Sonia Corbett Karkare
- Neuroscience Graduate Program, Emory University, Atlanta, GA, 30322, USA
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Hymavathy Balasubramanian
- Neuroscience Graduate Program, Emory University, Atlanta, GA, 30322, USA
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | | | - Jarildy Larimar Javier
- Neuroscience Graduate Program, Emory University, Atlanta, GA, 30322, USA
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Maha Rashid
- Neuroscience Graduate Program, Emory University, Atlanta, GA, 30322, USA
- Department of Biology, Emory University, Atlanta, GA, 30322, USA
| | - Malavika Murugan
- Neuroscience Graduate Program, Emory University, Atlanta, GA, 30322, USA.
- Department of Biology, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
23
|
Li DC, Hinton EA, Guo J, Knight KA, Sequeira MK, Wynne ME, Dighe NM, Gourley SL. Social experience in adolescence shapes prefrontal cortex structure and function in adulthood. Mol Psychiatry 2024; 29:2787-2798. [PMID: 38580810 PMCID: PMC11567502 DOI: 10.1038/s41380-024-02540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/07/2024]
Abstract
During adolescence, the prefrontal cortex (PFC) undergoes dramatic reorganization. PFC development is profoundly influenced by the social environment, disruptions to which may prime the emergence of psychopathology across the lifespan. We investigated the neurobehavioral consequences of isolation experienced in adolescence in mice, and in particular, the long-term consequences that were detectable even despite normalization of the social milieu. Isolation produced biases toward habit-like behavior at the expense of flexible goal seeking, plus anhedonic-like reward deficits. Behavioral phenomena were accompanied by neuronal dendritic spine over-abundance and hyper-excitability in the ventromedial PFC (vmPFC), which was necessary for the expression of isolation-induced habits and sufficient to trigger behavioral inflexibility in socially reared controls. Isolation activated cytoskeletal regulatory pathways otherwise suppressed during adolescence, such that repression of constituent elements prevented long-term isolation-induced neurosequelae. Altogether, our findings unveil an adolescent critical period and multi-model mechanism by which social experiences facilitate prefrontal cortical maturation.
Collapse
Affiliation(s)
- Dan C Li
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA, USA.
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
| | - Elizabeth A Hinton
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Jidong Guo
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Psychiatry and Behavioral sciences, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Michelle K Sequeira
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Meghan E Wynne
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Niharika M Dighe
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Shannon L Gourley
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Psychiatry and Behavioral sciences, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
24
|
Guo T, Feng Y, Zhou J, Meng L, Zhu X, Chen X, Xiao L, Feng L, Zhang L, Xiang YT, Zhao YJ, Wang G. Unveiling the Interplay Between Depressive Symptoms' Alleviation and Quality of Life Improvement in Major Depressive Disorder: A Network Analysis Based on Longitudinal Data. Neuropsychiatr Dis Treat 2024; 20:1641-1654. [PMID: 39228960 PMCID: PMC11370766 DOI: 10.2147/ndt.s462884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
Background Understanding the dynamic relationship between depressive symptoms and quality of life (QOL) is essential in improving long-term outcomes for patients with Major Depressive Disorder (MDD). While previous studies often relied on cross-sectional data, there is a pressing need for stronger evidence based on longitudinal data to better inform the development of effective clinical interventions. By focusing on key depressive symptoms, such interventions have the potential to ultimately enhance QOL in individuals with MDD. Methods This multi-center prospective study, conducted between 2016 and 2020, enrolled outpatients and inpatients diagnosed with MDD across twelve psychiatric hospitals in China. Longitudinal data on Patient Health Questionnaire - 9 (PHQ-9) and Quality of Life Enjoyment and Satisfaction Questionnaire-Short Form (Q-LES-Q-SF) was analyzed using an Extended Bayesian Information Criterion (EBIC) graphical least absolute shrinkage and selection operator (gLASSO) network model to explore the connections between depressive symptom changes and QOL changes. Flow network was applied to investigate relationships between individual symptom changes and overall QOL score change, as well as daily functional independence. Results This study included 818 participants with complete data after 8-week antidepressant treatment. Apart from the overlapping items from PHQ-9 and Q-LES-Q-SF, the three edges between "mood" (delta-QLES2) and "anhedonia" (delta-DEP1), between "physical health" (delta-QLES1) and "sleep problems" (delta-DEP3), and between "physical health" (delta-QLES1) and "sad mood" (delta-DEP2) were the most strong bridges between the cluster of depressive symptoms alleviation and the cluster of QOL change. "Anhedonia" (delta-DEP1), "sad mood" (delta-DEP2) and "loss of energy" (delta-DEP4) had the highest bridge strength between the alleviations of depressive symptoms and the total score change of Q-LES-Q-SF. Anhedonia had the greatest connection with participants' satisfaction with function in daily life. Conclusion This study highlighted the potential for developing highly effective interventions by targeting on central symptoms, thereby to ultimately improve QOL for patients with MDD.
Collapse
Affiliation(s)
- Tong Guo
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Yuan Feng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Jingjing Zhou
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Linghui Meng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Xuequan Zhu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Xu Chen
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Le Xiao
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Lei Feng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Ling Zhang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Yu-Tao Xiang
- Unit of Psychiatry, Department of Public Health and Medicinal Administration, Faculty of Health Sciences, Institute of Translational Medicine, University of Macau, Taipa, Macao SAR, People’s Republic of China
- Centre for Cognitive and Brain Sciences, University of Macau, TaipaMacao SAR, People’s Republic of China
| | - Yan-Jie Zhao
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Gang Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
25
|
Runyon K, Bui T, Mazanek S, Hartle A, Marschalko K, Howe WM. Distinct cholinergic circuits underlie discrete effects of reward on attention. Front Mol Neurosci 2024; 17:1429316. [PMID: 39268248 PMCID: PMC11390659 DOI: 10.3389/fnmol.2024.1429316] [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: 05/07/2024] [Accepted: 08/01/2024] [Indexed: 09/15/2024] Open
Abstract
Attention and reward are functions that are critical for the control of behavior, and massive multi-region neural systems have evolved to support the discrete computations associated with each. Previous research has also identified that attention and reward interact, though our understanding of the neural mechanisms that mediate this interplay is incomplete. Here, we review the basic neuroanatomy of attention, reward, and cholinergic systems. We then examine specific contexts in which attention and reward computations interact. Building on this work, we propose two discrete neural circuits whereby acetylcholine, released from cell groups located in different parts of the brain, mediates the impact of stimulus-reward associations as well as motivation on attentional control. We conclude by examining these circuits as a potential shared loci of dysfunction across diseases states associated with deficits in attention and reward.
Collapse
Affiliation(s)
- Kelly Runyon
- School of Neuroscience at Virginia Tech, Blacksburg, VA, United States
| | - Tung Bui
- School of Neuroscience at Virginia Tech, Blacksburg, VA, United States
| | - Sarah Mazanek
- School of Neuroscience at Virginia Tech, Blacksburg, VA, United States
| | - Alec Hartle
- School of Neuroscience at Virginia Tech, Blacksburg, VA, United States
| | - Katie Marschalko
- School of Neuroscience at Virginia Tech, Blacksburg, VA, United States
| | | |
Collapse
|
26
|
Luscher B, Jiang T, Feng M, Hutsell A. Sex-specific GABAergic microcircuits that switch vulnerability into resilience to stress and reverse the effects of chronic stress exposure. RESEARCH SQUARE 2024:rs.3.rs-4408723. [PMID: 39041032 PMCID: PMC11261964 DOI: 10.21203/rs.3.rs-4408723/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Clinical and preclinical studies have identified somatostatin (SST)-positive interneurons as key elements that regulate the vulnerability to stress-related psychiatric disorders. Conversely, disinhibition of SST neurons in mice results in resilience to the behavioral effects of chronic stress. Here we established a low-dose chronic chemogenetic protocol to map these changes in positively and negatively motivated behaviors to specific brain regions. AAV-hM3Dq mediated chronic activation of SST neurons in the prelimbic cortex (PLC) had antidepressant drug-like effects on anxiety- and anhedonia-related motivated behaviors in male but not female mice. Analogous manipulation of the ventral hippocampus (vHPC) had such effects in female but not male mice. Moreover, activation of SST neurons in the PLC of male and the vHPC of female mice resulted in stress resilience. Activation of SST neurons in the PLC reversed prior chronic stress-induced defects in motivated behavior in males but was ineffective in females. Conversely, activation of SST neurons in the vHPC reversed chronic stress-induced behavioral alterations in females but not males. Quantitation of c-Fos+ and FosB+ neurons in chronic stress-exposed mice revealed that chronic activation of SST neurons leads to a paradoxical increase in pyramidal cell activity. Collectively, these data demonstrate that GABAergic microcircuits driven by dendrite targeting interneurons enable sex- and brain-region-specific neural plasticity that promotes stress resilience and reverses stress-induced anxiety- and anhedonia-like motivated behavior. Our studies provide a mechanistic rationale for antidepressant efficacy of dendrite-targeting, low-potency GABAA receptor agonists, independent of sex and despite striking sex differences in the relevant brain substrates.
Collapse
|
27
|
Li Y, Lee SH, Yu C, Hsu LM, Wang TWW, Do K, Kim HJ, Shih YYI, Grill WM. Optogenetic fMRI reveals therapeutic circuits of subthalamic nucleus deep brain stimulation. Brain Stimul 2024; 17:947-957. [PMID: 39096961 PMCID: PMC11364984 DOI: 10.1016/j.brs.2024.07.022] [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: 04/12/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024] Open
Abstract
While deep brain stimulation (DBS) is widely employed for managing motor symptoms in Parkinson's disease (PD), its exact circuit mechanisms remain controversial. To identify the neural targets affected by therapeutic DBS in PD, we analyzed DBS-evoked whole brain activity in female hemi-parkinsonian rats using functional magnetic resonance imaging (fMRI). We delivered subthalamic nucleus (STN) DBS at various stimulation pulse repetition rates using optogenetics, allowing unbiased examination of cell-type specific STN feedforward neural activity. Unilateral optogenetic STN DBS elicited pulse repetition rate-dependent alterations of blood-oxygenation-level-dependent (BOLD) signals in SNr (substantia nigra pars reticulata), GP (globus pallidus), and CPu (caudate putamen). Notably, this modulation effectively ameliorated pathological circling behavior in animals expressing the kinetically faster Chronos opsin, but not in animals expressing ChR2. Furthermore, mediation analysis revealed that the pulse repetition rate-dependent behavioral rescue was significantly mediated by optogenetic DBS induced activity changes in GP and CPu, but not in SNr. This suggests that the activation of GP and CPu are critically involved in the therapeutic mechanisms of STN DBS.
Collapse
Affiliation(s)
- Yuhui Li
- Department of Biomedical Engineering, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Chunxiu Yu
- Department of Biomedical Engineering, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Wen W Wang
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Khoa Do
- Department of Biomedical Engineering, USA
| | - Hyeon-Joong Kim
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA.
| | - Warren M Grill
- Department of Biomedical Engineering, USA; Department of Electrical and Computer Engineering, USA; Department of Neurobiology, Duke University, Durham, NC, USA; Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
28
|
Goodwin NL, Choong JJ, Hwang S, Pitts K, Bloom L, Islam A, Zhang YY, Szelenyi ER, Tong X, Newman EL, Miczek K, Wright HR, McLaughlin RJ, Norville ZC, Eshel N, Heshmati M, Nilsson SRO, Golden SA. Simple Behavioral Analysis (SimBA) as a platform for explainable machine learning in behavioral neuroscience. Nat Neurosci 2024; 27:1411-1424. [PMID: 38778146 PMCID: PMC11268425 DOI: 10.1038/s41593-024-01649-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 04/12/2024] [Indexed: 05/25/2024]
Abstract
The study of complex behaviors is often challenging when using manual annotation due to the absence of quantifiable behavioral definitions and the subjective nature of behavioral annotation. Integration of supervised machine learning approaches mitigates some of these issues through the inclusion of accessible and explainable model interpretation. To decrease barriers to access, and with an emphasis on accessible model explainability, we developed the open-source Simple Behavioral Analysis (SimBA) platform for behavioral neuroscientists. SimBA introduces several machine learning interpretability tools, including SHapley Additive exPlanation (SHAP) scores, that aid in creating explainable and transparent behavioral classifiers. Here we show how the addition of explainability metrics allows for quantifiable comparisons of aggressive social behavior across research groups and species, reconceptualizing behavior as a sharable reagent and providing an open-source framework. We provide an open-source, graphical user interface (GUI)-driven, well-documented package to facilitate the movement toward improved automation and sharing of behavioral classification tools across laboratories.
Collapse
Affiliation(s)
- Nastacia L Goodwin
- Department of Biological Structure, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
- Center of Excellence in Neurobiology of Addiction, Pain and Emotion (NAPE), University of Washington, Seattle, WA, USA
| | - Jia J Choong
- Department of Biological Structure, University of Washington, Seattle, WA, USA
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Sophia Hwang
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Kayla Pitts
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Liana Bloom
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Aasiya Islam
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Yizhe Y Zhang
- Department of Biological Structure, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
- Center of Excellence in Neurobiology of Addiction, Pain and Emotion (NAPE), University of Washington, Seattle, WA, USA
| | - Eric R Szelenyi
- Department of Biological Structure, University of Washington, Seattle, WA, USA
- Center of Excellence in Neurobiology of Addiction, Pain and Emotion (NAPE), University of Washington, Seattle, WA, USA
| | - Xiaoyu Tong
- New York University Neuroscience Institute, New York, NY, USA
| | - Emily L Newman
- Department of Psychiatry, Harvard Medical School McLean Hospital, Belmont, MA, USA
| | - Klaus Miczek
- Department of Psychology, Tufts University, Medford, MA, USA
| | - Hayden R Wright
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, USA
| | - Ryan J McLaughlin
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, USA
| | | | - Neir Eshel
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Mitra Heshmati
- Department of Biological Structure, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
- Center of Excellence in Neurobiology of Addiction, Pain and Emotion (NAPE), University of Washington, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Simon R O Nilsson
- Department of Biological Structure, University of Washington, Seattle, WA, USA.
| | - Sam A Golden
- Department of Biological Structure, University of Washington, Seattle, WA, USA.
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
- Center of Excellence in Neurobiology of Addiction, Pain and Emotion (NAPE), University of Washington, Seattle, WA, USA.
| |
Collapse
|
29
|
Salabat D, Pourebrahimi A, Mayeli M, Cattarinussi G. The Therapeutic Role of Intermittent Theta Burst Stimulation in Schizophrenia: A Systematic Review and Meta-analysis. J ECT 2024; 40:78-87. [PMID: 38277616 DOI: 10.1097/yct.0000000000000972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
ABSTRACT Schizophrenia affects approximately 1% of the population worldwide. Multifactorial reasons, ranging from drug resistance to adverse effects of medications, have necessitated exploring further therapeutic options. Intermittent theta burst stimulation (iTBS) is a novel high-frequency form of transcranial magnetic stimulation, a safe procedure with minor adverse effects with faster and longer-lasting poststimulation effects with a potential role in treating symptoms; however, the exact target brain regions and symptoms are still controversial. Therefore, we aimed to systematically investigate the current literature regarding the therapeutic utilities of iTBS using Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines. Twelve studies were included among which 9 found iTBS effective to some degree. These studies targeted the dorsolateral prefrontal cortex and the midline cerebellum. We performed a random-effects meta-analysis on studies that compared the effects of iTBS on schizophrenia symptoms measured by the Positive and Negative Syndrome Scale (PANSS) to sham treatment. Our results showed no significant difference between iTBS and sham in PANSS positive and negative scores, but a trend-level difference in PANSS general scores ( k = 6, P = 0.07), and a significant difference in PANSS total scores ( k = 6, P = 0.03). Analysis of the studies targeting the dorsolateral prefrontal cortex showed improvement in PANSS negative scores ( k = 5, standardized mean difference = -0.83, P = 0.049), but not in PANSS positive scores. Moderators (intensity, pulse, quality, sessions) did not affect the results. However, considering the small number of studies included in this meta-analysis, future works are required to further explore the effects of these factors and also find optimum target regions for positive symptoms.
Collapse
|
30
|
Jüllig AK, Hebib S, Metzker H, Gruber E, Gruber O. Task-induced deactivation dysfunction during reward processing is associated with low self-esteem in a possible subtype of major depression. Brain Behav 2024; 14:e3545. [PMID: 38873863 DOI: 10.1002/brb3.3545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/10/2024] [Accepted: 05/02/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Low self-esteem is a frequent symptom in major depressive disorder (MDD). This functional magnetic resonance imaging study investigated whether MDD patients with low self-esteem show a distinct neural pathophysiology. Previous studies linked low self-esteem to reduced task-induced deactivation of the pregenual anterior cingulate cortex (pgACC) as a part of the default mode network, and to reduced connectivity between pgACC and reward system. Goya-Maldonado et al. identified an MDD subtype with pgACC and ventral striatal overactivations during reward processing. We hypothesized that this subtype might be characterized by low self-esteem. METHODS Eighty-three MDD patients performed the desire-reason dilemma task and completed the Rosenberg Self-Esteem Scale (RSES). Brain activity during bottom-up reward processing was regressed upon the RSES scores, controlling for depression severity measured by the Montgomery-Åsberg Depression Rating Scale. To corroborate the findings, we compared self-esteem scores between patient subgroups with impaired task-induced deactivation (n = 31) and with preserved task-induced deactivation (n = 31) of the pgACC. RESULTS Consistent with our a priori hypothesis, activity in a bilateral fronto-striatal network including pgACC and ventral striatum correlated negatively with RSES scores, also when controlling for depression severity. In the additional analysis, patients with impaired task-induced pgACC deactivation showed lower self-esteem (t (52.82) = -2.27; p = .027, d = 0.58) compared to those with preserved task-induced pgACC deactivation. CONCLUSIONS We conclude that low self-esteem in MDD patients is linked to a task-induced deactivation dysfunction of the pgACC. Our findings suggest that a previously described possible subtype of MDD with pgACC and ventral striatal overactivations during reward processing is clinically characterized by low self-esteem.
Collapse
Affiliation(s)
- Antonia K Jüllig
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, University Hospital Heidelberg, Heidelberg, Germany
| | - Sandi Hebib
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, University Hospital Heidelberg, Heidelberg, Germany
| | - Helena Metzker
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, University Hospital Heidelberg, Heidelberg, Germany
| | - Eva Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, University Hospital Heidelberg, Heidelberg, Germany
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
31
|
Spreen A, Alkhoury D, Walter H, Müller S. Optogenetic behavioral studies in depression research: A systematic review. iScience 2024; 27:109776. [PMID: 38726370 PMCID: PMC11079475 DOI: 10.1016/j.isci.2024.109776] [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: 07/27/2023] [Revised: 10/21/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Optogenetics has made substantial contributions to our understanding of the mechanistic underpinnings of depression. This systematic review employs quantitative analysis to investigate the impact of optogenetic stimulation in mice and rats on behavioral alterations in social interaction, sucrose consumption, and mobility. The review analyses optogenetic behavioral studies using standardized behavioral tests to detect behavioral changes induced via optogenetic stimulation in stressed or stress-naive mice and rats. Behavioral changes were evaluated as either positive, negative, or not effective. The analysis comprises the outcomes of 248 behavioral tests of 168 studies described in 37 articles, including negative and null results. Test outcomes were compared for each behavior, depending on the animal cohort, applied type of stimulation and the stimulated neuronal circuit and cell type. The presented synthesis contributes toward a comprehensive picture of optogenetic behavioral research in the context of depression.
Collapse
Affiliation(s)
- Anika Spreen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
- Experimental Biophysics, Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dana Alkhoury
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| | - Henrik Walter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| | - Sabine Müller
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| |
Collapse
|
32
|
Botterill JJ, Khlaifia A, Appings R, Wilkin J, Violi F, Premachandran H, Cruz-Sanchez A, Canella AE, Patel A, Zaidi SD, Arruda-Carvalho M. Dorsal peduncular cortex activity modulates affective behavior and fear extinction in mice. Neuropsychopharmacology 2024; 49:993-1006. [PMID: 38233571 PMCID: PMC11039686 DOI: 10.1038/s41386-024-01795-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024]
Abstract
The medial prefrontal cortex (mPFC) is critical to cognitive and emotional function and underlies many neuropsychiatric disorders, including mood, fear and anxiety disorders. In rodents, disruption of mPFC activity affects anxiety- and depression-like behavior, with specialized contributions from its subdivisions. The rodent mPFC is divided into the dorsomedial prefrontal cortex (dmPFC), spanning the anterior cingulate cortex (ACC) and dorsal prelimbic cortex (PL), and the ventromedial prefrontal cortex (vmPFC), which includes the ventral PL, infralimbic cortex (IL), and in some studies the dorsal peduncular cortex (DP) and dorsal tenia tecta (DTT). The DP/DTT have recently been implicated in the regulation of stress-induced sympathetic responses via projections to the hypothalamus. While many studies implicate the PL and IL in anxiety-, depression-like and fear behavior, the contribution of the DP/DTT to affective and emotional behavior remains unknown. Here, we used chemogenetics and optogenetics to bidirectionally modulate DP/DTT activity and examine its effects on affective behaviors, fear and stress responses in C57BL/6J mice. Acute chemogenetic activation of DP/DTT significantly increased anxiety-like behavior in the open field and elevated plus maze tests, as well as passive coping in the tail suspension test. DP/DTT activation also led to an increase in serum corticosterone levels and facilitated auditory fear extinction learning and retrieval. Activation of DP/DTT projections to the dorsomedial hypothalamus (DMH) acutely decreased freezing at baseline and during extinction learning, but did not alter affective behavior. These findings point to the DP/DTT as a new regulator of affective behavior and fear extinction in mice.
Collapse
Affiliation(s)
- Justin J Botterill
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Abdessattar Khlaifia
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Ryan Appings
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Jennifer Wilkin
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Francesca Violi
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Hanista Premachandran
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Arely Cruz-Sanchez
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S3G5, Canada
| | - Anna Elisabete Canella
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Ashutosh Patel
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - S Danyal Zaidi
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S3G5, Canada.
| |
Collapse
|
33
|
Zou Y, Tong C, Peng W, Qiu Y, Li J, Xia Y, Pei M, Zhang K, Li W, Xu M, Liang Z. Cell-type-specific optogenetic fMRI on basal forebrain reveals functional network basis of behavioral preference. Neuron 2024; 112:1342-1357.e6. [PMID: 38359827 DOI: 10.1016/j.neuron.2024.01.017] [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: 07/31/2023] [Revised: 12/12/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024]
Abstract
The basal forebrain (BF) is a complex structure that plays key roles in regulating various brain functions. However, it remains unclear how cholinergic and non-cholinergic BF neurons modulate large-scale functional networks and their relevance in intrinsic and extrinsic behaviors. With an optimized awake mouse optogenetic fMRI approach, we revealed that optogenetic stimulation of four BF neuron types evoked distinct cell-type-specific whole-brain BOLD activations, which could be attributed to BF-originated low-dimensional structural networks. Additionally, optogenetic activation of VGLUT2, ChAT, and PV neurons in the BF modulated the preference for locomotion, exploration, and grooming, respectively. Furthermore, we uncovered the functional network basis of the above BF-modulated behavioral preference through a decoding model linking the BF-modulated BOLD activation, low-dimensional structural networks, and behavioral preference. To summarize, we decoded the functional network basis of differential behavioral preferences with cell-type-specific optogenetic fMRI on the BF and provided an avenue for investigating mouse behaviors from a whole-brain view.
Collapse
Affiliation(s)
- Yijuan Zou
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 201602, China
| | - Chuanjun Tong
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 201602, China
| | - Wanling Peng
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yue Qiu
- Cardiac Intensive Care Center, Zhongshan Hospital, Fudan University Shanghai, Shanghai 200032, China
| | - Jiangxue Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Xia
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengchao Pei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kaiwei Zhang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weishuai Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Min Xu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Zhifeng Liang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 201602, China.
| |
Collapse
|
34
|
Adeyelu T, Vaughn T, Ogundele OM. VTA Excitatory Neurons Control Reward-driven Behavior by Modulating Infralimbic Cortical Firing. Neuroscience 2024; 548:50-68. [PMID: 38513762 DOI: 10.1016/j.neuroscience.2024.03.012] [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: 08/15/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
The functional dichotomy of anatomical regions of the medial prefrontal cortex (mPFC) has been tested with greater certainty in punishment-driven tasks, and less so in reward-oriented paradigms. In the infralimbic cortex (IL), known for behavioral suppression (STOP), tasks linked with reward or punishment are encoded through firing rate decrease or increase, respectively. Although the ventral tegmental area (VTA) is the brain region governing reward/aversion learning, the link between its excitatory neuron population and IL encoding of reward-linked behavioral expression is unclear. Here, we present evidence that IL ensembles use a population-based mechanism involving broad inhibition of principal cells at intervals when reward is presented or expected. The IL encoding mechanism was consistent across multiple sessions with randomized rewarded target sites. Most IL neurons exhibit FR (Firing Rate) suppression during reward acquisition intervals (T1), and subsequent exploration of previously rewarded targets when the reward is omitted (T2). Furthermore, FR suppression in putative IL ensembles persisted for intervals that followed reward-linked target events. Pairing VTA glutamate inhibition with reward acquisition events reduced the weight of reward-target association expressed as a lower affinity for previously rewarded targets. For these intervals, fewer IL neurons per mouse trial showed FR decrease and were accompanied by an increase in the percentage of units with no change in FR. Together, we conclude that VTA glutamate neurons are likely involved in establishing IL inhibition states that encode reward acquisition, and subsequent reward-target association.
Collapse
Affiliation(s)
- Tolulope Adeyelu
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, United States
| | - Tashonda Vaughn
- Department of Environmental Toxicology, College of Agriculture, Southern University A&M College, Baton Rouge, LA 70813, United States
| | - Olalekan M Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, United States.
| |
Collapse
|
35
|
Liu D, Zheng X, Hui Y, Xu Y, Du J, Du Z, Che Y, Wu F, Yu G, Zhang J, Gong X, Guo G. Lateral hypothalamus orexinergic projection to the medial prefrontal cortex modulates chronic stress-induced anhedonia but not anxiety and despair. Transl Psychiatry 2024; 14:149. [PMID: 38493173 PMCID: PMC10944479 DOI: 10.1038/s41398-024-02860-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Chronic stress-induced anxiodepression is a common health problem, however its potential neurocircuitry mechanism remains unclear. We used behavioral, patch-clamp electrophysiology, chemogenetic, and optogenetic approaches to clarify the response of the lateral hypothalamus (LH) and the medial prefrontal cortex (mPFC) to stress, confirmed the structural connections between the LH and mPFC, and investigated the role of the LH-mPFC pathway in chronic stress-induced anxiodepression symptoms. Unpredictable chronic mild stress (UCMS) caused anxiodepression-like behaviors, including anxiety, anhedonia, and despair behaviors. We discovered that the activity of the LH and mPFC was both increased after restraint stress (RS), a stressor of UCMS. Then we found that the orexinergic neurons in the LH predominantly project to the glutamatergic neurons in the mPFC, and the excitability of these neurons were increased after UCMS. In addition, overactivated LH orexinergic terminals in the mPFC induced anhedonia but not anxiety and despair behaviors in naive mice. Moreover, chemogenetically inhibited LH-mPFC orexinergic projection neurons and blocked the orexin receptors in the mPFC alleviated anhedonia but not anxiety and despair behaviors in UCMS-treated mice. Our study identified a new neurocircuit from LH orexinergic neurons to mPFC and revealed its role in regulating anhedonia in response to stress. Overactivation of LHOrx-mPFC pathway selectively mediated chronic stress-induced anhedonia. In normal mice, the LHOrx-mPFC pathway exhibits relatively low activity. However, after chronic stress, the activity of orexinergic neuron in LH is overactivated, leading to an increased release of orexin into the mPFC. This heightened orexin concentration results in increased excitability of the mPFC through OX1R and OX2R, consequently triggering anhedonia.
Collapse
Affiliation(s)
- Danlei Liu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Xuefeng Zheng
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
| | - Yuqing Hui
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yuanyuan Xu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
| | - Jinjiang Du
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
| | - Zean Du
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
| | - Yichen Che
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
| | - Fengming Wu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
| | - Guangyin Yu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China.
| | - Xiaobing Gong
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
36
|
Cerri DH, Albaugh DL, Walton LR, Katz B, Wang TW, Chao THH, Zhang W, Nonneman RJ, Jiang J, Lee SH, Etkin A, Hall CN, Stuber GD, Shih YYI. Distinct neurochemical influences on fMRI response polarity in the striatum. Nat Commun 2024; 15:1916. [PMID: 38429266 PMCID: PMC10907631 DOI: 10.1038/s41467-024-46088-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 02/13/2024] [Indexed: 03/03/2024] Open
Abstract
The striatum, known as the input nucleus of the basal ganglia, is extensively studied for its diverse behavioral roles. However, the relationship between its neuronal and vascular activity, vital for interpreting functional magnetic resonance imaging (fMRI) signals, has not received comprehensive examination within the striatum. Here, we demonstrate that optogenetic stimulation of dorsal striatal neurons or their afferents from various cortical and subcortical regions induces negative striatal fMRI responses in rats, manifesting as vasoconstriction. These responses occur even with heightened striatal neuronal activity, confirmed by electrophysiology and fiber-photometry. In parallel, midbrain dopaminergic neuron optogenetic modulation, coupled with electrochemical measurements, establishes a link between striatal vasodilation and dopamine release. Intriguingly, in vivo intra-striatal pharmacological manipulations during optogenetic stimulation highlight a critical role of opioidergic signaling in generating striatal vasoconstriction. This observation is substantiated by detecting striatal vasoconstriction in brain slices after synthetic opioid application. In humans, manipulations aimed at increasing striatal neuronal activity likewise elicit negative striatal fMRI responses. Our results emphasize the necessity of considering vasoactive neurotransmission alongside neuronal activity when interpreting fMRI signal.
Collapse
Affiliation(s)
- Domenic H Cerri
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel L Albaugh
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Lindsay R Walton
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brittany Katz
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Wen Wang
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Weiting Zhang
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Randal J Nonneman
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jing Jiang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sung-Ho Lee
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amit Etkin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Alto Neuroscience, Los Altos, CA, USA
| | - Catherine N Hall
- Sussex Neuroscience, University of Sussex, Falmer, United Kingdom
- School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Garret D Stuber
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Biomedical Research Imaging Center, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Neurology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
37
|
Li Y, Lee SH, Yu C, Hsu LM, Wang TWW, Do K, Kim HJ, Shih YYI, Grill WM. Optogenetic fMRI reveals therapeutic circuits of subthalamic nucleus deep brain stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581627. [PMID: 38464010 PMCID: PMC10925223 DOI: 10.1101/2024.02.22.581627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
While deep brain stimulation (DBS) is widely employed for managing motor symptoms in Parkinson's disease (PD), its exact circuit mechanisms remain controversial. To identify the neural targets affected by therapeutic DBS in PD, we analyzed DBS-evoked whole brain activity in female hemi-parkinsonian rats using function magnetic resonance imaging (fMRI). We delivered subthalamic nucleus (STN) DBS at various stimulation pulse repetition rates using optogenetics, allowing unbiased examinations of cell-type specific STN feed-forward neural activity. Unilateral STN optogenetic stimulation elicited pulse repetition rate-dependent alterations of blood-oxygenation-level-dependent (BOLD) signals in SNr (substantia nigra pars reticulata), GP (globus pallidus), and CPu (caudate putamen). Notably, these manipulations effectively ameliorated pathological circling behavior in animals expressing the kinetically faster Chronos opsin, but not in animals expressing ChR2. Furthermore, mediation analysis revealed that the pulse repetition rate-dependent behavioral rescue was significantly mediated by optogenetically induced activity changes in GP and CPu, but not in SNr. This suggests that the activation of GP and CPu are critically involved in the therapeutic mechanisms of STN DBS.
Collapse
|
38
|
Beaulieu M. Capturing wild animal welfare: a physiological perspective. Biol Rev Camb Philos Soc 2024; 99:1-22. [PMID: 37635128 DOI: 10.1111/brv.13009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/29/2023]
Abstract
Affective states, such as emotions, are presumably widespread across the animal kingdom because of the adaptive advantages they are supposed to confer. However, the study of the affective states of animals has thus far been largely restricted to enhancing the welfare of animals managed by humans in non-natural contexts. Given the diversity of wild animals and the variable conditions they can experience, extending studies on animal affective states to the natural conditions that most animals experience will allow us to broaden and deepen our general understanding of animal welfare. Yet, this same diversity makes examining animal welfare in the wild highly challenging. There is therefore a need for unifying theoretical frameworks and methodological approaches that can guide researchers keen to engage in this promising research area. The aim of this article is to help advance this important research area by highlighting the central relationship between physiology and animal welfare and rectify its apparent oversight, as revealed by the current scientific literature on wild animals. Moreover, this article emphasises the advantages of including physiological markers to assess animal welfare in the wild (e.g. objectivity, comparability, condition range, temporality), as well as their concomitant limitations (e.g. only access to peripheral physiological markers with complex relationships with affective states). Best-practice recommendations (e.g. replication and multifactorial approaches) are also provided to allow physiological markers to be used most effectively and appropriately when assessing the welfare of animals in their natural habitat. This review seeks to provide the foundation for a new and distinct research area with a vast theoretical and applied potential: wild animal welfare physiology.
Collapse
Affiliation(s)
- Michaël Beaulieu
- Wild Animal Initiative, 5123 W 98th St, 1204, Minneapolis, MN, 55437, USA
| |
Collapse
|
39
|
Costello H, Husain M, Roiser JP. Apathy and Motivation: Biological Basis and Drug Treatment. Annu Rev Pharmacol Toxicol 2024; 64:313-338. [PMID: 37585659 DOI: 10.1146/annurev-pharmtox-022423-014645] [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] [Indexed: 08/18/2023]
Abstract
Apathy is a disabling syndrome associated with poor functional outcomes that is common across a broad range of neurological and psychiatric conditions. Currently, there are no established therapies specifically for the condition, and safe and effective treatments are urgently needed. Advances in the understanding of motivation and goal-directed behavior in humans and animals have shed light on the cognitive and neurobiological mechanisms contributing to apathy, providing an important foundation for the development of new treatments. Here, we review the cognitive components, neural circuitry, and pharmacology of apathy and motivation, highlighting converging evidence of shared transdiagnostic mechanisms. Though no pharmacological treatments have yet been licensed, we summarize trials of existing and novel compounds to date, identifying several promising candidates for clinical use and avenues of future drug development.
Collapse
Affiliation(s)
- Harry Costello
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom;
| | - Masud Husain
- Nuffield Department of Clinical Neurosciences and Department of Experimental Psychology, Oxford University, Oxford, United Kingdom
| | - Jonathan P Roiser
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom;
| |
Collapse
|
40
|
Li M, Yang G. A mesocortical glutamatergic pathway modulates neuropathic pain independent of dopamine co-release. Nat Commun 2024; 15:643. [PMID: 38245542 PMCID: PMC10799877 DOI: 10.1038/s41467-024-45035-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
Dysfunction in the mesocortical pathway, connecting the ventral tegmental area (VTA) to the prefrontal cortex, has been implicated in chronic pain. While extensive research has focused on the role of dopamine, the contribution of glutamatergic signaling in pain modulation remains unknown. Using in vivo calcium imaging, we observe diminished VTA glutamatergic activity targeting the prelimbic cortex (PL) in a mouse model of neuropathic pain. Optogenetic activation of VTA glutamatergic terminals in the PL alleviates neuropathic pain, whereas inhibiting these terminals in naïve mice induces pain-like responses. Importantly, this pain-modulating effect is independent of dopamine co-release, as demonstrated by CRISPR/Cas9-mediated gene deletion. Furthermore, we show that VTA neurons primarily project to excitatory neurons in the PL, and their activation restores PL outputs to the anterior cingulate cortex, a key region involved in pain processing. These findings reveal a distinct mesocortical glutamatergic pathway that critically modulates neuropathic pain independent of dopamine signaling.
Collapse
Affiliation(s)
- Miao Li
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
41
|
Mandino F, Vujic S, Grandjean J, Lake EMR. Where do we stand on fMRI in awake mice? Cereb Cortex 2024; 34:bhad478. [PMID: 38100331 PMCID: PMC10793583 DOI: 10.1093/cercor/bhad478] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 12/17/2023] Open
Abstract
Imaging awake animals is quickly gaining traction in neuroscience as it offers a means to eliminate the confounding effects of anesthesia, difficulties of inter-species translation (when humans are typically imaged while awake), and the inability to investigate the full range of brain and behavioral states in unconscious animals. In this systematic review, we focus on the development of awake mouse blood oxygen level dependent functional magnetic resonance imaging (fMRI). Mice are widely used in research due to their fast-breeding cycle, genetic malleability, and low cost. Functional MRI yields whole-brain coverage and can be performed on both humans and animal models making it an ideal modality for comparing study findings across species. We provide an analysis of 30 articles (years 2011-2022) identified through a systematic literature search. Our conclusions include that head-posts are favorable, acclimation training for 10-14 d is likely ample under certain conditions, stress has been poorly characterized, and more standardization is needed to accelerate progress. For context, an overview of awake rat fMRI studies is also included. We make recommendations that will benefit a wide range of neuroscience applications.
Collapse
Affiliation(s)
- Francesca Mandino
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, United States
| | - Stella Vujic
- Department of Computer Science, Yale University, New Haven, CT 06520, United States
| | - Joanes Grandjean
- Donders Institute for Brain, Behaviour, and Cognition, Radboud University, Nijmegen, The Netherlands
- Department for Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evelyn M R Lake
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, United States
| |
Collapse
|
42
|
Xiang S, Jia T, Xie C, Zhu Z, Cheng W, Schumann G, Robbins TW, Feng J. Fractionation of neural reward processing into independent components by novel decoding principle. Neuroimage 2023; 284:120463. [PMID: 37989457 DOI: 10.1016/j.neuroimage.2023.120463] [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: 04/16/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 11/23/2023] Open
Abstract
How to retrieve latent neurobehavioural processes from complex neurobiological signals is an important yet unresolved challenge. Here, we develop a novel approach, orthogonal-Decoding multi-Cognitive Processes (DeCoP), to reveal underlying latent neurobehavioural processing and show that its performance is superior to traditional non-orthogonal decoding in terms of both false inference and robustness. Processing value and salience information are two fundamental but mutually confounded pathways of reward reinforcement essential for decision making. During reward/punishment anticipation, we applied DeCoP to decode brain-wide responses into spatially overlapping, yet functionally independent, evaluation and readiness processes, which are modulated differentially by meso‑limbic vs nigro-striatal dopamine systems. Using DeCoP, we further demonstrated that most brain regions only encoded abstract information but not the exact input, except for dorsal anterior cingulate cortex and insula. Furthermore, we anticipate our novel analytical principle to be applied generally in decoding multiple latent neurobehavioral processes and thus advance both the design and hypothesis testing for cognitive tasks.
Collapse
Affiliation(s)
- Shitong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), China
| | - Tianye Jia
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), China; Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, SE5 8AF, United Kingdom; Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Chao Xie
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), China
| | - Zhichao Zhu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), China
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), China
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Department of Psychiatry and Psychotherapy, Centre for Population Neuroscience and Precision Medicine (PONS), CCM, Charite Universitaetsmedizin, Berlin, Germany
| | - Trevor W Robbins
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), China; Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), China; Department of Computer Science, University of Warwick, Coventry, United Kingdom; School of Mathematical Sciences and Centre for Computational Systems Biology, Fudan University, Shanghai, China.
| |
Collapse
|
43
|
Zhu J, Jiao Y, Chen R, Wang XH, Han Y. Aberrant dynamic and static functional connectivity of the striatum across specific low-frequency bands in patients with autism spectrum disorder. Psychiatry Res Neuroimaging 2023; 336:111749. [PMID: 37977097 DOI: 10.1016/j.pscychresns.2023.111749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 10/06/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Dysfunctions of the striatum have been repeatedly observed in autism spectrum disorder (ASD). However, previous studies have explored the static functional connectivity (sFC) of the striatum in a single frequency band, ignoring the dynamics and frequency specificity of brain FC. Therefore, we investigated the dynamic FC (dFC) and sFC of the striatum in the slow-4 (0.027-0.073 Hz) and slow-5 (0.01-0.027 Hz) frequency bands. METHODS Data of 47 ASD patients and 47 typically developing (TD) controls were obtained from the Autism Brain Imaging Data Exchange (ABIDE) database. A seed-based approach was used to compute the dFC and sFC. Then, a two-sample t-test was performed. For regions showing abnormal sFC and dFC, we performed clinical correlation analysis and constructed support vector machine (SVM) models. RESULTS The middle frontal gyrus (MFG), precuneus, and medial superior frontal gyrus (mPFC) showed both dynamic and static alterations. The reduced striatal dFC in the right MFG was associated with autism symptoms. The dynamic‒static FC model had a great performance in ASD classification, with 95.83 % accuracy. CONCLUSIONS The striatal dFC and sFC were altered in ASD, which were frequency specific. Examining brain activity using dynamic and static FC provides a comprehensive view of brain activity.
Collapse
Affiliation(s)
- Junsa Zhu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Yun Jiao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China; Network Information Center, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China.
| | - Ran Chen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Xun-Heng Wang
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Yunyan Han
- Public Health School of Dalian Medical University, Dalian 116000, China
| |
Collapse
|
44
|
Ayala-Rodríguez JD, García-Colunga J. Maternal separation modifies spontaneous synaptic activity in the infralimbic cortex of stress-resilient male rats. PLoS One 2023; 18:e0294151. [PMID: 37943747 PMCID: PMC10635473 DOI: 10.1371/journal.pone.0294151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Glutamate and GABA signaling systems are necessary to maintain proper function of the central nervous system through excitation/inhibition (E/I) balance. Alteration of this balance in the medial prefrontal cortex (mPFC), as an effect of early-life stress, may lead to the development of anxiety and depressive disorders. Few studies exist in the infralimbic division of the mPFC to understand the effect of early-life stress at different ages, which is the purpose of the present work. Newborn Sprague Dawley male rats were subjected to maternal separation (MS) for two weeks. First, tests measuring anxiety- and depression-like behaviors were performed on adolescent and adult rats subjected to MS (MS-rats). Then, to establish a relationship with behavioral results, electrophysiological recordings were performed in neurons of the infralimbic cortex in acute brain slices of infant, adolescent, and adult rats. In the behavioral tests, there were no significant differences in MS-rats compared to control rats at any age. Moreover, MS had no effect on the passive membrane properties nor neuronal excitability in the infralimbic cortex, whereas spontaneous synaptic activity in infralimbic neurons was altered. The frequency of spontaneous glutamatergic synaptic events increased in infant MS-rats, whereas in adolescent MS-rats both the frequency and the amplitude of spontaneous GABAergic events increased without any effect on glutamatergic synaptic responses. In adult MS-rats, these two parameters decreased in spontaneous GABAergic synaptic events, whereas only the frequency of glutamatergic events decreased. These data suggest that rats subjected to MS did not exhibit behavioral changes and presented an age-dependent E/I imbalance in the infralimbic cortex, possibly due to differential changes in neurotransmitter release and/or receptor expression.
Collapse
Affiliation(s)
- Jesús David Ayala-Rodríguez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Jesús García-Colunga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| |
Collapse
|
45
|
Weber F, Hong J, Lozano D, Beier K, Chung S. Prefrontal Cortical Regulation of REM Sleep. RESEARCH SQUARE 2023:rs.3.rs-1417511. [PMID: 37886570 PMCID: PMC10602053 DOI: 10.21203/rs.3.rs-1417511/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Rapid-eye-movement (REM) sleep is accompanied by intense cortical activity, underlying its wake-like electroencephalogram (EEG). The neural activity inducing REM sleep is thought to originate from subcortical circuits in brainstem and hypothalamus. However, whether cortical neurons can also trigger REM sleep has remained unknown. Here, we show in mice that the medial prefrontal cortex (mPFC) strongly promotes REM sleep. Bidirectional optogenetic manipulations demonstrate that excitatory mPFC neurons promote REM sleep through their projections to the lateral hypothalamus (LH) and regulate phasic events, reflected in accelerated EEG theta oscillations and increased eye-movement density during REM sleep. Calcium imaging reveals that the majority of LH-projecting mPFC neurons are maximally activated during REM sleep and a subpopulation is recruited during phasic theta accelerations. Our results delineate a cortico-hypothalamic circuit for the top-down control of REM sleep and identify a critical role of the mPFC in regulating phasic events during REM sleep.
Collapse
|
46
|
Flores-García M, Rizzo A, Garçon-Poca MZ, Fernández-Dueñas V, Bonaventura J. Converging circuits between pain and depression: the ventral tegmental area as a therapeutic hub. Front Pharmacol 2023; 14:1278023. [PMID: 37849731 PMCID: PMC10577189 DOI: 10.3389/fphar.2023.1278023] [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: 08/15/2023] [Accepted: 09/25/2023] [Indexed: 10/19/2023] Open
Abstract
Chronic pain and depression are highly prevalent pathologies and cause a major socioeconomic burden to society. Chronic pain affects the emotional state of the individuals suffering from it, while depression worsens the prognosis of chronic pain patients and may diminish the effectiveness of pain treatments. There is a high comorbidity rate between both pathologies, which might share overlapping mechanisms. This review explores the evidence pinpointing a role for the ventral tegmental area (VTA) as a hub where both pain and emotional processing might converge. In addition, the feasibility of using the VTA as a possible therapeutic target is discussed. The role of the VTA, and the dopaminergic system in general, is highly studied in mood disorders, especially in deficits in reward-processing and motivation. Conversely, the VTA is less regarded where it concerns the study of central mechanisms of pain and its mood-associated consequences. Here, we first outline the brain circuits involving central processing of pain and mood disorders, focusing on the often-understudied role of the dopaminergic system and the VTA. Next, we highlight the state-of-the-art findings supporting the emergence of the VTA as a link where both pathways converge. Thus, we envision a promising part for the VTA as a putative target for innovative therapeutic approaches to treat chronic pain and its effects on mood. Finally, we emphasize the urge to develop and use animal models where both pain and depression-like symptoms are considered in conjunction.
Collapse
Affiliation(s)
- Montse Flores-García
- Unitat de Farmacologia, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat, Catalonia, Spain
| | - Arianna Rizzo
- Unitat de Farmacologia, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat, Catalonia, Spain
| | - Maria Zelai Garçon-Poca
- Unitat de Farmacologia, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat, Catalonia, Spain
| | - Víctor Fernández-Dueñas
- Unitat de Farmacologia, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat, Catalonia, Spain
| | - Jordi Bonaventura
- Unitat de Farmacologia, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat, Catalonia, Spain
| |
Collapse
|
47
|
Hong J, Lozano DE, Beier KT, Chung S, Weber F. Prefrontal cortical regulation of REM sleep. Nat Neurosci 2023; 26:1820-1832. [PMID: 37735498 DOI: 10.1038/s41593-023-01398-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/28/2023] [Indexed: 09/23/2023]
Abstract
Rapid eye movement (REM) sleep is accompanied by intense cortical activity, underlying its wake-like electroencephalogram. The neural activity inducing REM sleep is thought to originate from subcortical circuits in brainstem and hypothalamus. However, whether cortical neurons can also trigger REM sleep has remained unknown. Here we show in mice that the medial prefrontal cortex (mPFC) strongly promotes REM sleep. Bidirectional optogenetic manipulations demonstrate that excitatory mPFC neurons promote REM sleep through their projections to the lateral hypothalamus and regulate phasic events, reflected in accelerated electroencephalogram theta oscillations and increased eye movement density during REM sleep. Calcium imaging reveals that the majority of lateral hypothalamus-projecting mPFC neurons are maximally activated during REM sleep and a subpopulation is recruited during phasic theta accelerations. Our results delineate a cortico-hypothalamic circuit for the top-down control of REM sleep and identify a critical role of the mPFC in regulating phasic events during REM sleep.
Collapse
Affiliation(s)
- Jiso Hong
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - David E Lozano
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Shinjae Chung
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Franz Weber
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Managò F, Scheggia D, Pontillo M, Mereu M, Mastrogiacomo R, Udayan G, Valentini P, Tata MC, Weinberger DR, Weickert CS, Pompa PP, De Luca MA, Vicari S, Papaleo F. Dopaminergic signalling and behavioural alterations by Comt-Dtnbp1 genetic interaction and their clinical relevance. Br J Pharmacol 2023; 180:2514-2531. [PMID: 37218669 DOI: 10.1111/bph.16147] [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: 06/13/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND AND PURPOSE Cognitive and motor functions are modulated by dopaminergic signalling, which is shaped by several genetic factors. The biological effects of single genetic variants might differ depending on epistatic interactions that can be functionally multi-directional and non-linear. EXPERIMENTAL APPROACH We performed behavioural and neurochemical assessments in genetically modified mice and behavioural assessments and genetic screening in human patients with 22q11.2 deletion syndrome (22q11.2DS). KEY RESULTS Here, we confirm a genetic interaction between the Comt (catechol-O-methyltransferase, human orthologue: COMT) and Dtnbp1 (dystrobrevin binding protein 1, alias dysbindin, human orthologue: DTNBP1) genes that modulate cortical and striatal dopaminergic signalling in a manner not predictable by the effects of each single gene. In mice, Comt-by-Dtnbp1 concomitant reduction leads to a hypoactive mesocortical and a hyperactive mesostriatal dopamine pathway, associated with specific cognitive abnormalities. Like mice, in subjects with the 22q11.2DS (characterized by COMT hemideletion and dopamine alterations), COMT-by-DTNBP1 concomitant reduction was associated with analogous cognitive disturbances. We then developed an easy and inexpensive colourimetric kit for the genetic screening of common COMT and DTNBP1 functional genetic variants for clinical application. CONCLUSIONS AND IMPLICATIONS These findings illustrate an epistatic interaction of two dopamine-related genes and their functional effects, supporting the need to address genetic interaction mechanisms at the base of complex behavioural traits.
Collapse
Affiliation(s)
- Francesca Managò
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Diego Scheggia
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maria Pontillo
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maddalena Mereu
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Rosa Mastrogiacomo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Gayatri Udayan
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Paola Valentini
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Cynthia S Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Pier Paolo Pompa
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Maria A De Luca
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stefano Vicari
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
49
|
Xiao J, Provenza NR, Asfouri J, Myers J, Mathura RK, Metzger B, Adkinson JA, Allawala AB, Pirtle V, Oswalt D, Shofty B, Robinson ME, Mathew SJ, Goodman WK, Pouratian N, Schrater PR, Patel AB, Tolias AS, Bijanki KR, Pitkow X, Sheth SA. Decoding Depression Severity From Intracranial Neural Activity. Biol Psychiatry 2023; 94:445-453. [PMID: 36736418 PMCID: PMC10394110 DOI: 10.1016/j.biopsych.2023.01.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/09/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Disorders of mood and cognition are prevalent, disabling, and notoriously difficult to treat. Fueling this challenge in treatment is a significant gap in our understanding of their neurophysiological basis. METHODS We recorded high-density neural activity from intracranial electrodes implanted in depression-relevant prefrontal cortical regions in 3 human subjects with severe depression. Neural recordings were labeled with depression severity scores across a wide dynamic range using an adaptive assessment that allowed sampling with a temporal frequency greater than that possible with typical rating scales. We modeled these data using regularized regression techniques with region selection to decode depression severity from the prefrontal recordings. RESULTS Across prefrontal regions, we found that reduced depression severity is associated with decreased low-frequency neural activity and increased high-frequency activity. When constraining our model to decode using a single region, spectral changes in the anterior cingulate cortex best predicted depression severity in all 3 subjects. Relaxing this constraint revealed unique, individual-specific sets of spatiospectral features predictive of symptom severity, reflecting the heterogeneous nature of depression. CONCLUSIONS The ability to decode depression severity from neural activity increases our fundamental understanding of how depression manifests in the human brain and provides a target neural signature for personalized neuromodulation therapies.
Collapse
Affiliation(s)
- Jiayang Xiao
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas; Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| | - Nicole R Provenza
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Joseph Asfouri
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas
| | - John Myers
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Raissa K Mathura
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Brian Metzger
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Joshua A Adkinson
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | | | - Victoria Pirtle
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Denise Oswalt
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Ben Shofty
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Meghan E Robinson
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Sanjay J Mathew
- Department of Psychiatry, Baylor College of Medicine, Houston, Texas
| | - Wayne K Goodman
- Department of Psychiatry, Baylor College of Medicine, Houston, Texas
| | - Nader Pouratian
- Department of Neurological Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Paul R Schrater
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota; Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | - Ankit B Patel
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas; Department of Electrical and Computer Engineering, Rice University, Houston, Texas; Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, Texas
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas; Department of Electrical and Computer Engineering, Rice University, Houston, Texas; Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, Texas
| | - Kelly R Bijanki
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Xaq Pitkow
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas; Department of Electrical and Computer Engineering, Rice University, Houston, Texas; Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, Texas
| | - Sameer A Sheth
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
50
|
Jang DC, Choi S, Chung G, Kim SK. Global Cerebral Ischemia-induced Depression Accompanies Alteration of Neuronal Excitability in the Infralimbic Cortex Layer 2/3 Pyramidal Neurons. Exp Neurobiol 2023; 32:302-312. [PMID: 37749930 PMCID: PMC10569139 DOI: 10.5607/en23017] [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: 06/21/2023] [Revised: 07/19/2023] [Accepted: 08/30/2023] [Indexed: 09/27/2023] Open
Abstract
Cerebral ischemia can lead to a range of sequelae, including depression. The pathogenesis of depression involves neuronal change of the medial prefrontal cortex (mPFC). However, how cerebral ischemia-induced changes manifest across subregions and layers of the mPFC is not well understood. In this study, we induced cerebral ischemia in mice via transient bilateral common carotid artery occlusion (tBCCAO) and observed depressive-like behavior. Using whole-cell patch clamp recording, we identified changes in the excitability of pyramidal neurons in the prelimbic cortex (PL) and infralimbic cortex (IL), the subregions of mPFC. Compared to sham control mice, tBCCAO mice showed significantly reduced neuronal excitability in IL layer 2/3 but not layer 5 pyramidal neurons, accompanied by increased rheobase current and decreased input resistance. In contrast, no changes were observed in the excitability of PL layer 2/3 and layer 5 pyramidal neurons. Our results provide a new direction for studying the pathogenesis of depression following ischemic damage by showing that cerebral ischemia induces subregion- and layer-specific changes in the mPFC pyramidal neurons.
Collapse
Affiliation(s)
- Dong Cheol Jang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Seunghwan Choi
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|