1
|
Lin FV, Simmons JM, Turnbull A, Zuo Y, Conwell Y, Wang KH. Cross-Species Framework for Emotional Well-Being and Brain Aging: Lessons From Behavioral Neuroscience. JAMA Psychiatry 2025:2833240. [PMID: 40332879 DOI: 10.1001/jamapsychiatry.2025.0581] [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] [Indexed: 05/08/2025]
Abstract
Importance Emotional well-being (EWB) is an emerging therapeutic target for managing and preventing symptoms associated with Alzheimer disease and related dementias (ADRD). However, more research is needed to establish causal inferences between brain changes, EWB, and behavioral changes observed in typical aging and ADRD. Observations This article presents a framework for using a cross-species behavioral neuroscience approach to study EWB and brain aging, adopting a well-established biobehavioral model that highlights the reciprocal roles of brain changes, EWB, and ADRD symptoms. First, the challenges and opportunities in this field are reviewed. Then, a practical solution to improve comparability between animal and human studies is proposed. Conclusions and Relevance The goal is to draw comprehensive parallels and distinctions that could enhance the understanding of the mechanisms linking brain aging, EWB, and ADRD symptomatic disturbances across different species.
Collapse
Affiliation(s)
- F Vankee Lin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Janine M Simmons
- Office of Behavioral and Social Sciences Research, National Institutes of Health, Bethesda, Maryland
| | - Adam Turnbull
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Yi Zuo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
| | - Yeates Conwell
- Department of Psychiatry, University of Rochester, Rochester, New York
| | - Kuan Hong Wang
- Department of Neuroscience, University of Rochester, Rochester, New York
| |
Collapse
|
2
|
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
|
3
|
Porta-de-la-Riva M, Gonzalez AC, Sanfeliu-Cerdán N, Karimi S, Malaiwong N, Pidde A, Morales-Curiel LF, Fernandez P, González-Bolívar S, Hurth C, Krieg M. Neural engineering with photons as synaptic transmitters. Nat Methods 2023; 20:761-769. [PMID: 37024651 DOI: 10.1038/s41592-023-01836-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 03/03/2023] [Indexed: 04/08/2023]
Abstract
Neuronal computation is achieved through connections of individual neurons into a larger network. To expand the repertoire of endogenous cellular communication, we developed a synthetic, photon-assisted synaptic transmission (PhAST) system. PhAST is based on luciferases and channelrhodopsins that enable the transmission of a neuronal state across space, using photons as neurotransmitters. PhAST overcomes synaptic barriers and rescues the behavioral deficit of a glutamate mutant with conditional, calcium-triggered photon emission between two neurons of the Caenorhabditis elegans nociceptive avoidance circuit. To demonstrate versatility and flexibility, we generated de novo synaptic transmission between two unconnected cells in a sexually dimorphic neuronal circuit, suppressed endogenous nocifensive response through activation of an anion channelrhodopsin and switched attractive to aversive behavior in an olfactory circuit. Finally, we applied PhAST to dissect the calcium dynamics of the temporal pattern generator in a motor circuit for ovipositioning. In summary, we established photon-based synaptic transmission that facilitates the modification of animal behavior.
Collapse
Affiliation(s)
| | | | | | - Shadi Karimi
- Institut de Ciències Fotòniques, Castelldefels, Spain
| | | | | | | | | | | | - Cedric Hurth
- Institut de Ciències Fotòniques, Castelldefels, Spain
| | - Michael Krieg
- Institut de Ciències Fotòniques, Castelldefels, Spain.
| |
Collapse
|
4
|
Teal LB, Ingram SM, Bubser M, McClure E, Jones CK. The Evolving Role of Animal Models in the Discovery and Development of Novel Treatments for Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:37-99. [PMID: 36928846 DOI: 10.1007/978-3-031-21054-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Elliott McClure
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
5
|
Walsh JJ, Christoffel DJ, Malenka RC. Neural circuits regulating prosocial behaviors. Neuropsychopharmacology 2023; 48:79-89. [PMID: 35701550 PMCID: PMC9700801 DOI: 10.1038/s41386-022-01348-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/09/2022] [Accepted: 05/17/2022] [Indexed: 11/09/2022]
Abstract
Positive, prosocial interactions are essential for survival, development, and well-being. These intricate and complex behaviors are mediated by an amalgamation of neural circuit mechanisms working in concert. Impairments in prosocial behaviors, which occur in a large number of neuropsychiatric disorders, result from disruption of the coordinated activity of these neural circuits. In this review, we focus our discussion on recent findings that utilize modern approaches in rodents to map, monitor, and manipulate neural circuits implicated in a variety of prosocial behaviors. We highlight how modulation by oxytocin, serotonin, and dopamine of excitatory and inhibitory synaptic transmission in specific brain regions is critical for regulation of adaptive prosocial interactions. We then describe how recent findings have helped elucidate pathophysiological mechanisms underlying the social deficits that accompany neuropsychiatric disorders. We conclude by discussing approaches for the development of more efficacious and targeted therapeutic interventions to ameliorate aberrant prosocial behaviors.
Collapse
Affiliation(s)
- Jessica J Walsh
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27514, USA.
- Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA.
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27514, USA.
| | - Daniel J Christoffel
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27514, USA
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, 27514, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA.
| |
Collapse
|
6
|
A Miniaturized Closed-Loop Optogenetic Brain Stimulation Device. ELECTRONICS 2022. [DOI: 10.3390/electronics11101591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This paper presents a tetherless and miniaturized closed-loop optogenetic brain stimulation device, designed as a back mountable device for laboratory mice. The device has the ability to sense the biomarkers corresponding to major depressive disorder (MDD) from local field potential (LFP), and produces a feedback signal to control the closed-loop operation after on-device processing of the sensed signals. MDD is a chronic neurological disorder and there are still many unanswered questions about the underlying neurological mechanisms behind its occurrence. Along with other brain stimulation paradigms, optogenetics has recently proved effective in the study of MDD. Most of these experiments have used tethered and connected devices. However, the use of tethered devices in optogenetic brain stimulation experiments has the drawback of hindering the free movement of the laboratory animal subjects undergoing stimulation. To address this issue, the proposed device is small, light-weight, untethered, and back-mountable. The device consists of: (i) an optrode which houses an electrode for collecting neural signals, an optical source for delivering light stimulations, and a temperature sensor for monitoring the temperature increase at the stimulation site, (ii) a neural sensor for acquisition and pre-processing of the neural signals to obtain LFP signals in the frequency range of 4 to 200 Hz, as electrophysiological biomarkers of MDD (iii) a classifier for classification of the signal into four classes: normal, abnormal alpha, abnormal theta, and abnormal gamma oscillations, (iv) a control algorithm to select stimulation parameters based on the input class, and (v) a stimulator for generating light stimulations. The design, implementation, and evaluation of the device are presented, and the results are discussed. The neural sensor and the stimulator are circular in shape with a radius of 8 mm. Pre-recorded neural signals from the mouse hippocampus are used for the evaluation of the device.
Collapse
|
7
|
Kochanski RB, Slavin KV. The future perspectives of psychiatric neurosurgery. PROGRESS IN BRAIN RESEARCH 2022; 270:211-228. [PMID: 35396029 DOI: 10.1016/bs.pbr.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The future of psychiatric neurosurgery can be viewed from two separate perspectives: the immediate future and the distant future. Both show promise, but the treatment strategy for mental diseases and the technology utilized during these separate periods will likely differ dramatically. It can be expected that the initial advancements will be built upon progress of neuroimaging and stereotactic targeting while surgical technology becomes adapted to patient-specific symptomatology and structural/functional imaging parameters. This individualized approach has already begun to show significant promise when applied to deep brain stimulation for treatment-resistant depression and obsessive-compulsive disorder. If effectiveness of these strategies is confirmed by well designed, double-blind, placebo-controlled clinical studies, further technological advances will continue into the distant future, and will likely involve precise neuromodulation at the cellular level, perhaps using wireless technology with or without closed-loop design. This approach, being theoretically less invasive and carrying less risk, may ultimately propel psychiatric neurosurgery to the forefront in the treatment algorithm of mental illness. Despite prominent development of non-invasive therapeutic options, such as stereotactic radiosurgery or transcranial magnetic resonance-guided focused ultrasound, chances are there will still be a need in surgical management of patients with most intractable psychiatric conditions.
Collapse
Affiliation(s)
- Ryan B Kochanski
- Neurosurgery, Methodist Healthcare System, San Antonio, TX, United States
| | - Konstantin V Slavin
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL, United States; Neurology Service, Jesse Brown Veterans Administration Medical Center, Chicago, IL, United States.
| |
Collapse
|
8
|
Spadini S, Ferro M, Lamanna J, Malgaroli A. Activity-based anorexia animal model: a review of the main neurobiological findings. J Eat Disord 2021; 9:123. [PMID: 34600568 PMCID: PMC8487535 DOI: 10.1186/s40337-021-00481-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The genesis of anorexia nervosa (AN), a severe eating disorder with a pervasive effect on many brain functions such as attention, emotions, reward processing, cognition and motor control, has not yet been understood. Since our current knowledge of the genetic aspects of AN is limited, we are left with a large and diversified number of biological, psychological and environmental risk factors, called into question as potential triggers of this chronic condition with a high relapse rate. One of the most valid and used animal models for AN is the activity-based anorexia (ABA), which recapitulates important features of the human condition. This model is generated from naïve rodents by a self-motivated caloric restriction, where a fixed schedule food delivery induces spontaneous increased physical activity. AIM In this review, we sought to provide a summary of the experimental research conducted using the ABA model in the pursuit of potential neurobiological mechanism(s) underlying AN. METHOD The experimental work presented here includes evidence for neuroanatomical and neurophysiological changes in several brain regions as well as for the dysregulation of specific neurochemical synaptic and neurohormonal pathways. RESULTS The most likely hypothesis for the mechanism behind the development of the ABA phenotype relates to an imbalance of the neural circuitry that mediates reward processing. Evidence collected here suggests that ABA animals show a large set of alterations, involving regions whose functions extend way beyond the control of reward mechanisms and eating habits. Hence, we cannot exclude a primary role of these alterations from a mechanistic theory of ABA induction. CONCLUSIONS These findings are not sufficient to solve such a major enigma in neuroscience, still they could be used to design ad hoc further experimental investigation. The prospect is that, since treatment of AN is still challenging, the ABA model could be more effectively used to shed light on the complex AN neurobiological framework, thus supporting the future development of therapeutic strategies but also the identification of biomarkers and diagnostic tools. Anorexia Nervosa (AN) is a severe eating disorder with a dramatic effect on many functions of our brain, such as attention, emotions, cognition and motion control. Since our current knowledge of the genetic aspects behind the development of AN is still limited, many biological, psychological and environmental factors must be taken into account as potential triggers of this condition. One of the most valid animal models for studying AN is the activity-based anorexia (ABA). In this model, rodents spontaneously limit food intake and start performing increased physical activity on a running wheel, a result of the imposition of a fixed time schedule for food delivery. In this review, we provide a detailed summary of the experimental research conducted using the ABA model, which includes extended evidence for changes in the anatomy and function of the brain of ABA rodents. The hope is that such integrated view will support the design of future experiments that will shed light on the complex brain mechanisms behind AN. Such advanced knowledge is crucial to find new, effective strategies for both the early diagnosis of AN and for its treatment.
Collapse
Affiliation(s)
- Sara Spadini
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy
| | - Mattia Ferro
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy
- Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy.
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
9
|
Lost in translation: no effect of repeated optogenetic cortico-striatal stimulation on compulsivity in rats. Transl Psychiatry 2021; 11:315. [PMID: 34031365 PMCID: PMC8144623 DOI: 10.1038/s41398-021-01448-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 11/30/2022] Open
Abstract
The orbitofrontal cortex-ventromedial striatum (OFC-VMS) circuitry is widely believed to drive compulsive behavior. Hyperactivating this pathway in inbred mice produces excessive and persistent self-grooming, which has been considered a model for human compulsivity. We aimed to replicate these findings in outbred rats, where there are few reliable compulsivity models. Male Long-Evans rats implanted with optical fibers into VMS and with opsins delivered into OFC received optical stimulation at parameters that produce OFC-VMS plasticity and compulsive grooming in mice. We then evaluated rats for compulsive self-grooming at six timepoints: before, during, immediately after, and 1 h after each stimulation, 1 and 2 weeks after the ending of a 6-day stimulation protocol. To further test for effects of OFC-VMS hyperstimulation, we ran animals in three standard compulsivity assays: marble burying, nestlet shredding, and operant attentional set-shifting. OFC-VMS stimulation did not increase self-grooming or induce significant changes in nestlet shredding, marble burying, or set-shifting in rats. Follow-on evoked potential studies verified that the stimulation protocol altered OFC-VMS synaptic weighting. In sum, although we induced physiological changes in the OFC-VMS circuitry, we could not reproduce in a strongly powered study in rats a model of compulsive behavior previously reported in mice. This suggests possible limitations to translation of mouse findings to species higher on the phylogenetic chain.
Collapse
|
10
|
Dissecting neural mechanisms of prosocial behaviors. Curr Opin Neurobiol 2020; 68:9-14. [PMID: 33278639 DOI: 10.1016/j.conb.2020.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022]
Abstract
Prosocial behaviors are essential for group cooperation, which enrich life experience and enhance survival. These complex behaviors are governed by intricate interactions between numerous neural circuits functioning in concert. Impairments in prosocial interactions result from disruptions of this coordinated brain activity and are a prominent feature of several pathological conditions including autism spectrum disorder, depression and addiction. Here we highlight recent studies that use advanced techniques to anatomically map, monitor and manipulate neural circuits that influence prosocial behavior. These recent findings provide important clues to unravel the complexities of the neural mechanisms that mediate prosocial interactions and offer insights into new strategies for the treatment of aberrant social behavior.
Collapse
|
11
|
Neuroanatomic and Functional Neuroimaging Findings. Curr Top Behav Neurosci 2020; 48:173-196. [PMID: 33040316 DOI: 10.1007/7854_2020_174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The search for brain morphology findings that could explain behavioral disorders has gone through a long path in the history of psychiatry. With the advance of brain imaging technology, studies have been able to identify brain morphology and neural circuits associated with the pathophysiology of mental illnesses, such as bipolar disorders (BD). Promising results have also shown the potential of neuroimaging findings in the identification of outcome predictors and response to treatment among patients with BD. In this chapter, we present brain imaging structural and functional findings associated with BD, as well as their hypothesized relationship with the pathophysiological aspects of that condition and their potential clinical applications.
Collapse
|
12
|
Kim S, Kim H, Park D, Kim J, Hong J, Kim JS, Jung H, Kim D, Cheong E, Ko J, Um JW. Loss of IQSEC3 Disrupts GABAergic Synapse Maintenance and Decreases Somatostatin Expression in the Hippocampus. Cell Rep 2020; 30:1995-2005.e5. [PMID: 32049026 DOI: 10.1016/j.celrep.2020.01.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/28/2019] [Accepted: 01/16/2020] [Indexed: 12/31/2022] Open
Abstract
Gephyrin interacts with various GABAergic synaptic proteins to organize GABAergic synapse development. Among the multitude of gephyrin-binding proteins is IQSEC3, a recently identified component at GABAergic synapses that acts through its ADP ribosylation factor-guanine nucleotide exchange factor (ARF-GEF) activity to orchestrate GABAergic synapse formation. Here, we show that IQSEC3 knockdown (KD) reduced GABAergic synaptic density in vivo, suggesting that IQSEC3 is required for GABAergic synapse maintenance in vivo. We further show that IQSEC3 KD in the dentate gyrus (DG) increases seizure susceptibility and triggers selective depletion of somatostatin (SST) peptides in the DG hilus in an ARF-GEP activity-dependent manner. Strikingly, selective introduction of SST into SST interneurons in DG-specific IQSEC3-KD mice reverses GABAergic synaptic deficits. Thus, our data suggest that IQSEC3 is required for linking gephyrin-GABAA receptor complexes with ARF-dependent pathways to prevent aberrant, runaway excitation and thereby contributes to the integrity of SST interneurons and proper GABAergic synapse maintenance.
Collapse
Affiliation(s)
- Seungjoon Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Hyeonho Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Dongseok Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Jinhu Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Joohyeon Hong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jae Seong Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Hyeji Jung
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Dongwook Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea.
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea; Core Protein Resources Center, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea.
| |
Collapse
|
13
|
van Kruining D, Luo Q, van Echten-Deckert G, Mielke MM, Bowman A, Ellis S, Oliveira TG, Martinez-Martinez P. Sphingolipids as prognostic biomarkers of neurodegeneration, neuroinflammation, and psychiatric diseases and their emerging role in lipidomic investigation methods. Adv Drug Deliv Rev 2020; 159:232-244. [PMID: 32360155 PMCID: PMC7665829 DOI: 10.1016/j.addr.2020.04.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/21/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023]
Abstract
Lipids play an important role in neurodegeneration, neuroinflammation, and psychiatric disorders and an imbalance in sphingolipid levels is associated with disease. Although early diagnosis and intervention of these disorders would clearly have favorable long-term outcomes, no diagnostic tests currently exist that can accurately identify people at risk. Reliable prognostic biomarkers that are easily accessible would be beneficial to determine therapy and treatment response in clinical trials. Recent advances in lipidomic investigation methods have greatly progressed the knowledge of sphingolipids in neurodegenerative and psychiatric disorders over the past decades although more longitudinal studies are needed to understand its exact role in these disorders to be used as potential tools in the clinic. In this review, we give an overview of the current knowledge of sphingolipids in neurodegenerative and psychiatric disorders and explore recent advances in investigation methods. Finally, the potential of sphingolipid metabolism products and signaling molecules as potential biomarkers for diagnosis, prognostic, or surrogate markers of treatment response is discussed.
Collapse
Affiliation(s)
- Daan van Kruining
- Division of Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Qian Luo
- Division of Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Gerhild van Echten-Deckert
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, Bonn, Germany
| | - Michelle M Mielke
- Department of Health Sciences Research and Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Andrew Bowman
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, the Netherlands
| | - Shane Ellis
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, the Netherlands
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), ICVS/3B's, School of Medicine, University of Minho, Braga, Portugal
| | - Pilar Martinez-Martinez
- Division of Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
14
|
Abstract
Monkeys are a premier model organism for neuroscience research. Activity in the central nervous systems of monkeys can be recorded and manipulated while they perform complex perceptual, motor, or cognitive tasks. Conventional techniques for manipulating neural activity in monkeys are too coarse to address many of the outstanding questions in primate neuroscience, but optogenetics holds the promise to overcome this hurdle. In this article, we review the progress that has been made in primate optogenetics over the past 5 years. We emphasize the use of gene regulatory sequences in viral vectors to target specific neuronal types, and we present data on vectors that we engineered to target parvalbumin-expressing neurons. We conclude with a discussion of the utility of optogenetics for treating sensorimotor hearing loss and Parkinson's disease, areas of translational neuroscience in which monkeys provide unique leverage for basic science and medicine.
Collapse
|
15
|
Frequency-specific effects of low-intensity rTMS can persist for up to 2 weeks post-stimulation: A longitudinal rs-fMRI/MRS study in rats. Brain Stimul 2019; 12:1526-1536. [PMID: 31296402 DOI: 10.1016/j.brs.2019.06.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 06/23/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Evidence suggests that repetitive transcranial magnetic stimulation (rTMS), a non-invasive neuromodulation technique, alters resting brain activity. Despite anecdotal evidence that rTMS effects wear off, there are no reports of longitudinal studies, even in humans, mapping the therapeutic duration of rTMS effects. OBJECTIVE Here, we investigated the longitudinal effects of repeated low-intensity rTMS (LI-rTMS) on healthy rodent resting-state networks (RSNs) using resting-state functional MRI (rs-fMRI) and on sensorimotor cortical neurometabolite levels using proton magnetic resonance spectroscopy (MRS). METHODS Sprague-Dawley rats received 10 min LI-rTMS daily for 15 days (10 Hz or 1 Hz stimulation, n = 9 per group). MRI data were acquired at baseline, after seven days and after 14 days of daily stimulation and at two more timepoints up to three weeks post-cessation of daily stimulation. RESULTS 10 Hz stimulation increased RSN connectivity and GABA, glutamine, and glutamate levels. 1 Hz stimulation had opposite but subtler effects, resulting in decreased RSN connectivity and glutamine levels. The induced changes decreased to baseline levels within seven days following stimulation cessation in the 10 Hz group but were sustained for at least 14 days in the 1 Hz group. CONCLUSION Overall, our study provides evidence of long-term frequency-specific effects of LI-rTMS. Additionally, the transient connectivity changes following 10 Hz stimulation suggest that current treatment protocols involving this frequency may require ongoing "top-up" stimulation sessions to maintain therapeutic effects.
Collapse
|
16
|
Blackburn TP. Depressive disorders: Treatment failures and poor prognosis over the last 50 years. Pharmacol Res Perspect 2019; 7:e00472. [PMID: 31065377 PMCID: PMC6498411 DOI: 10.1002/prp2.472] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/11/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Depression like many diseases is pleiotropic but unlike cancer and Alzheimer's disease for example, is still largely stigmatized and falls into the dark shadows of human illness. The failure of depression to be in the spotlight for successful treatment options is inherent in the complexity of the disease(s), flawed clinical diagnosis, overgeneralization of the illness, inadequate and biased clinical trial design, restrictive and biased inclusion/exclusion criteria, lack of approved/robust biomarkers, expensive imaging technology along with few advances in neurobiological hypotheses in decades. Clinical trial studies summitted to the regulatory agencies (FDA/EMA) for approval, have continually failed to show significant differences between active and placebo. For decades, we have acknowledged this failure, despite vigorous debated by all stakeholders to provide adequate answers to this escalating problem, with only a few new antidepressants approved in the last 20 years with equivocal efficacy, little improvement in side effects or onset of efficacy. It is also clear that funding and initiatives for mental illness lags far behind other life-treating diseases. Thus, it is no surprise we have not achieved much success in the last 50 years in treating depression, but we are accountable for the many failures and suboptimal treatment. This review will therefore critically address where we have failed and how future advances in medical science offers a glimmer of light for the patient and aid our future understanding of the neurobiology and pathophysiology of the disease, enabling transformative therapies for the treatment of depressive disorders.
Collapse
|
17
|
Lamanna J, Sulpizio S, Ferro M, Martoni R, Abutalebi J, Malgaroli A. Behavioral assessment of activity-based-anorexia: how cognition can become the drive wheel. Physiol Behav 2019; 202:1-7. [DOI: 10.1016/j.physbeh.2019.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/17/2019] [Accepted: 01/19/2019] [Indexed: 12/19/2022]
|
18
|
Farrell MR, Schoch H, Mahler SV. Modeling cocaine relapse in rodents: Behavioral considerations and circuit mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 2018; 87:33-47. [PMID: 29305936 PMCID: PMC6034989 DOI: 10.1016/j.pnpbp.2018.01.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/18/2017] [Accepted: 01/02/2018] [Indexed: 12/29/2022]
Abstract
Addiction is a chronic relapsing disorder, in that most addicted individuals who choose to quit taking drugs fail to maintain abstinence in the long-term. Relapse is especially likely when recovering addicts encounter risk factors like small "priming" doses of drug, stress, or drug-associated cues and locations. In rodents, these same factors reinstate cocaine seeking after a period of abstinence, and extensive preclinical work has used priming, stress, or cue reinstatement models to uncover brain circuits underlying cocaine reinstatement. Here, we review common rat models of cocaine relapse, and discuss how specific features of each model influence the neural circuits recruited during reinstated drug seeking. To illustrate this point, we highlight the surprisingly specific roles played by ventral pallidum subcircuits in cocaine seeking reinstated by either cocaine-associated cues, or cocaine itself. One goal of such studies is to identify, and eventually to reverse the specific circuit activity that underlies the inability of some humans to control their drug use. Based on preclinical findings, we posit that circuit activity in humans also differs based on the triggers that precipitate craving and relapse, and that associated neural responses could help predict the triggers most likely to elicit relapse in a given person. If so, examining circuit activity could facilitate diagnosis of subgroups of addicted people, allowing individualized treatment based on the most problematic risk factors.
Collapse
Affiliation(s)
- Mitchell R Farrell
- Department of Neurobiology & Behavior, University of California, 1203 McGaugh Hall, Irvine, United States
| | - Hannah Schoch
- Department of Neurobiology & Behavior, University of California, 1203 McGaugh Hall, Irvine, United States
| | - Stephen V Mahler
- Department of Neurobiology & Behavior, University of California, 1203 McGaugh Hall, Irvine, United States.
| |
Collapse
|
19
|
Robinson E. Psychopharmacology: From serendipitous discoveries to rationale design, but what next? Brain Neurosci Adv 2018; 2:2398212818812629. [PMID: 32166162 PMCID: PMC7058199 DOI: 10.1177/2398212818812629] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Indexed: 12/15/2022] Open
Abstract
Psychopharmacology really developed as a discipline from the mid-20th century with the discovery of a number of new classes of psychoactive drugs which could modify behaviour. These drugs were discovered as a consequence of clinical observations of patients, often being treated for other conditions. These serendipitous discoveries were the start of an era of drug development which has led to the antidepressants, antipsychotics, anxiolytics and mood stabilisers used today. Subsequent research focused on understanding why these drugs were effective, and used this information to develop a second generation of drugs that were more selective for their therapeutic targets, and therefore had reduced side effects and improved safety and tolerability. After a period of decline in new discoveries and withdrawal of the majority of the major pharmaceutical companies from active development programmes in psychiatry, new avenues are emerging fuelling renewed interest in this area.
Collapse
Affiliation(s)
- Emma Robinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
20
|
Nicholson JR, Sommer B. The research domain criteria framework in drug discovery for neuropsychiatric diseases: focus on negative valence. Brain Neurosci Adv 2018; 2:2398212818804030. [PMID: 32166151 PMCID: PMC7058263 DOI: 10.1177/2398212818804030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/24/2018] [Indexed: 12/22/2022] Open
Abstract
Drug discovery, particularly in the field of central nervous system, has had very limited success in the last few decades. A likely contributor is the poor translation between preclinical and clinical phases. The Research Domain Criteria of the National Institutes of Mental Health is a framework which aims to identify new ways of classifying mental illnesses that are based on observable behaviour and neurobiological measures, and to provide a guiding and evolving framework to improve the translation from preclinical to clinical research. At the core of the Research Domain Criteria approach is the assumption that the dimensional constructs described can be assessed across different units of analysis, thus enabling a more precise quantitative understanding of their neurobiological underpinnings, increasing the likelihood of identifying new and effective therapeutic approaches. In the present review, we discuss how the Research Domain Criteria can be applied to drug discovery with the domain Negative Valence, construct Potential Threat (‘Anxiety’) as an example. We will discuss the evidence supporting the utility of the Research Domain Criteria approach and evaluate how close we are to achieving a common thread of translational research from gene to self-report.
Collapse
Affiliation(s)
- Janet R Nicholson
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Bernd Sommer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
21
|
Lenz M, Galanis C, Kleidonas D, Fellenz M, Deller T, Vlachos A. Denervated mouse dentate granule cells adjust their excitatory but not inhibitory synapses following in vitro entorhinal cortex lesion. Exp Neurol 2018; 312:1-9. [PMID: 30401642 DOI: 10.1016/j.expneurol.2018.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/16/2018] [Accepted: 10/23/2018] [Indexed: 01/04/2023]
Abstract
Neurons adjust their synaptic strength in a homeostatic manner following changes in network activity and connectivity. While this form of plasticity has been studied in detail for excitatory synapses, homeostatic plasticity of inhibitory synapses remains not well-understood. In the present study, we employed entorhinal cortex lesion (ECL) of organotypic entorhino-hippocampal tissue cultures to test for homeostatic changes in GABAergic neurotransmission onto partially denervated dentate granule cells. Using single and paired whole-cell patch-clamp recordings, as well as immunostainings for synaptic markers, we find that excitatory synaptic strength is robustly increased 3 days post lesion (dpl), whereas GABAergic neurotransmission is not changed after denervation. Even under conditions of pharmacological inhibition of glutamatergic neurotransmission, which prevents neurons to compensate for the loss of input via excitatory synaptic scaling, down-scaling of GABAergic synapses does not emerge 3 days after denervation. We conclude that granule cells maintain structural and functional properties of GABAergic synapses even in the face of substantial changes in network connectivity. Hence, alterations in inhibitory neurotransmission, as seen in pathological brain states, may not simply reflect a homeostatic response to disconnection.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany; Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt 60590, Germany
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Meike Fellenz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt 60590, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt 60590, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.
| |
Collapse
|
22
|
Camporeze B, Manica BA, Bonafé GA, Ferreira JJC, Diniz AL, de Oliveira CTP, Mathias Junior LR, de Aguiar PHP, Ortega MM. Optogenetics: the new molecular approach to control functions of neural cells in epilepsy, depression and tumors of the central nervous system. Am J Cancer Res 2018; 8:1900-1918. [PMID: 30416844 PMCID: PMC6220144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/14/2018] [Indexed: 06/09/2023] Open
Abstract
The optogenetic tools have been described as valuable techniques to study neural activity through light stimulation, as well as potential neuromodulator approaches in the management of several central nervous system (CNS) diseases. Since the first bacteriorhodopsin protein described as a single-component light-activated regulator of transmembrane ion flow description, in 1980's, the focus has been on channel proteins for neurobiology; however, the advances in engineering techniques showed involvement changes in cellular biological behavior in several types of proteins involved in cell cytoskeleton regulation, motility and gene expression. Although the use of this technology has been published in many papers, a question still remains regarding real results and potential clinical applicability in CNS diseases, as well as the publications scarcity that systematically analyses the published results. Lastly, the aim of this review is to discuss the experimental results, molecular mechanisms and potential clinical applications of optogenetic tools in epilepsy and depression treatment, as well as its applicability in the treatment of CNS tumors.
Collapse
Affiliation(s)
- Bruno Camporeze
- Postgraduate Program in Health Science, Laboratory of Cellular and Molecular Biology and Bioactive Compounds, São Francisco University (USF)Bragança Paulista-SP, Brazil
- Postgraduate Program in Health Science, Department of Neurosurgery, Institute of Medical Assistance of The State Public Servant (IAMSPE)São Paulo-SP, Brazil
| | - Bruno Alcântara Manica
- Departament of Neurology, Medical School University Pontifical University Catholic of São Paulo (PUCSP)Sorocaba-SP, Brazil
| | - Gabriel Alves Bonafé
- Postgraduate Program in Health Science, Laboratory of Cellular and Molecular Biology and Bioactive Compounds, São Francisco University (USF)Bragança Paulista-SP, Brazil
| | | | - Aurélio Lourenço Diniz
- Departament of Neurology, Medical School University Pontifical University Catholic of São Paulo (PUCSP)Sorocaba-SP, Brazil
| | | | | | - Paulo Henrique Pires de Aguiar
- Postgraduate Program in Health Science, Department of Neurosurgery, Institute of Medical Assistance of The State Public Servant (IAMSPE)São Paulo-SP, Brazil
- Departament of Neurology, Medical School University Pontifical University Catholic of São Paulo (PUCSP)Sorocaba-SP, Brazil
- Departament of Neurosurgery, Hospital Santa PaulaSão Paulo-SP, Brazil
- Department of Research and Innovation, Laboratory of Cellular and Molecular Biology, Medical School of ABC (FMABC)Santo André-SP, Brazil
| | - Manoela Marques Ortega
- Postgraduate Program in Health Science, Laboratory of Cellular and Molecular Biology and Bioactive Compounds, São Francisco University (USF)Bragança Paulista-SP, Brazil
| |
Collapse
|
23
|
Bilge MT, Gosai AK, Widge AS. Deep Brain Stimulation in Psychiatry: Mechanisms, Models, and Next-Generation Therapies. Psychiatr Clin North Am 2018; 41:373-383. [PMID: 30098651 PMCID: PMC6092041 DOI: 10.1016/j.psc.2018.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Deep brain stimulation has preliminary evidence of clinical efficacy, but has been difficult to develop into a robust therapy, in part because its mechanisms are incompletely understood. We review evidence from movement and psychiatric disorder studies, with an emphasis on how deep brain stimulation changes brain networks. From this, we argue for a network-oriented approach to future deep brain stimulation studies. That network approach requires methods for identifying patients with specific circuit/network deficits. We describe how dimensional approaches to diagnoses may aid that identification. We discuss the use of network/circuit biomarkers to develop self-adjusting "closed loop" systems.
Collapse
Affiliation(s)
- Mustafa Taha Bilge
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Boston, MA 02129, USA
| | - Aishwarya K Gosai
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Boston, MA 02129, USA
| | - Alik S Widge
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Boston, MA 02129, USA; Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
24
|
Ben-Shaanan T, Schiller M, Rolls A. Studying brain-regulation of immunity with optogenetics and chemogenetics; A new experimental platform. Brain Behav Immun 2017; 65:1-8. [PMID: 27890661 DOI: 10.1016/j.bbi.2016.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023] Open
Abstract
The interactions between the brain and the immune system are bidirectional. Nevertheless, we have far greater understanding of how the immune system affects the brain than how the brain affects immunity. New technological developments such as optogenetics and chemogenetics (using DREADDs; Designer Receptors Exclusively Activated by Designer Drugs) can bridge this gap in our understanding, as they enable an unprecedented mechanistic and systemic analysis of the communication between the brain and the immune system. In this review, we discuss new experimental approaches for revealing neuronal circuits that can participate in regulation of immunity. In addition, we discuss methods, specifically optogenetics and chemogenetics, that enable targeted neuronal manipulation to reveal how different brain regions affect immunity. We describe how these techniques can be used as an experimental platform to address fundamental questions in psychoneuroimmunology and to understand how neuronal circuits associate with different psychological states can affect physiology.
Collapse
Affiliation(s)
- Tamar Ben-Shaanan
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Maya Schiller
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Asya Rolls
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
25
|
Bitzenhofer SH, Ahlbeck J, Hanganu-Opatz IL. Methodological Approach for Optogenetic Manipulation of Neonatal Neuronal Networks. Front Cell Neurosci 2017; 11:239. [PMID: 28848399 PMCID: PMC5554786 DOI: 10.3389/fncel.2017.00239] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/31/2017] [Indexed: 11/13/2022] Open
Abstract
Coordinated patterns of electrical activity are critical for the functional maturation of neuronal networks, yet their interrogation has proven difficult in the developing brain. Optogenetic manipulations strongly contributed to the mechanistic understanding of network activation in the adult brain, but difficulties to specifically and reliably express opsins at neonatal age hampered similar interrogation of developing circuits. Here, we introduce a protocol that enables to control the activity of specific neuronal populations by light, starting from early postnatal development. We show that brain area-, layer- and cell type-specific expression of opsins by in utero electroporation (IUE), as exemplified for the medial prefrontal cortex (PFC) and hippocampus (HP), permits the manipulation of neuronal activity in vitro and in vivo. Both individual and population responses to different patterns of light stimulation are monitored by extracellular multi-site recordings in the medial PFC of neonatal mice. The expression of opsins via IUE provides a flexible approach to disentangle the cellular mechanism underlying early rhythmic network activity, and to elucidate the role of early neuronal activity for brain maturation, as well as its contribution to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sebastian H Bitzenhofer
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-EppendorfHamburg, Germany
| | - Joachim Ahlbeck
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-EppendorfHamburg, Germany
| | - Ileana L Hanganu-Opatz
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-EppendorfHamburg, Germany
| |
Collapse
|
26
|
Jiang J, Cui H, Rahmouni K. Optogenetics and pharmacogenetics: principles and applications. Am J Physiol Regul Integr Comp Physiol 2017; 313:R633-R645. [PMID: 28794102 DOI: 10.1152/ajpregu.00091.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/18/2017] [Accepted: 08/05/2017] [Indexed: 12/29/2022]
Abstract
Remote and selective spatiotemporal control of the activity of neurons to regulate behavior and physiological functions has been a long-sought goal in system neuroscience. Identification and subsequent bioengineering of light-sensitive ion channels (e.g., channelrhodopsins, halorhodopsin, and archaerhodopsins) from the bacteria have made it possible to use light to artificially modulate neuronal activity, namely optogenetics. Recent advance in genetics has also allowed development of novel pharmacological tools to selectively and remotely control neuronal activity using engineered G protein-coupled receptors, which can be activated by otherwise inert drug-like small molecules such as the designer receptors exclusively activated by designer drug, a form of chemogenetics. The cutting-edge optogenetics and pharmacogenetics are powerful tools in neuroscience that allow selective and bidirectional modulation of the activity of defined populations of neurons with unprecedented specificity. These novel toolboxes are enabling significant advances in deciphering how the nervous system works and its influence on various physiological processes in health and disease. Here, we discuss the fundamental elements of optogenetics and chemogenetics approaches and some of the applications that yielded significant advances in various areas of neuroscience and beyond.
Collapse
Affiliation(s)
- Jingwei Jiang
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - Huxing Cui
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa; and.,Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa; .,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa; and.,Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
27
|
Keshavan MS, Lawler AN, Nasrallah HA, Tandon R. New drug developments in psychosis: Challenges, opportunities and strategies. Prog Neurobiol 2017; 152:3-20. [PMID: 27519538 PMCID: PMC5362348 DOI: 10.1016/j.pneurobio.2016.07.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 07/11/2016] [Indexed: 02/06/2023]
Abstract
All currently approved drugs for schizophrenia work mainly by dopaminergic antagonism. While they are efficacious for psychotic symptoms, their efficacy is limited for negative symptoms and cognitive deficits which underlie the substantive disability in this illness. Recent insights into the biological basis of schizophrenia, especially in relation to non-dopaminergic mechanisms, have raised the efforts to find novel and effective drug targets, though with relatively little success thus far. Potential impediments to novel drug discovery include the continued use of symptom based disease definitions which leads to etiological and pathophysiological heterogeneity, lack of valid preclinical models for drug testing, and design limitations in clinical trials. These roadblocks can be addressed by (i) characterizing trans-diagnostic, translational pathophysiological dimensions as potential treatment targets, (ii) efficiency, accountability and, transparency in approaches to the clinical trials process, and (iii) leveraging recent advances in genetics and in vitro phenotypes. Accomplishing these goals is urgent given the significant unmet needs in the pharmacological treatment of schizophrenia. As this happens, it is imperative that clinicians employ optimal dosing, measurement-based care, and other best practices in utilizing existing treatments to optimize outcomes for their patients today.
Collapse
Affiliation(s)
- Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Massachusetts Mental Health Center, Harvard Medical School, United States.
| | - Ashley N Lawler
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Massachusetts Mental Health Center, Harvard Medical School, United States
| | - Henry A Nasrallah
- Department of Neurology & Psychiatry, St Louis University, United States
| | - Rajiv Tandon
- Department of Psychiatry, University of Florida, Gainsville, Florida. and the North FL/South Georgia Veterans' Administration Medical Center, Gainesville, FL 32610, United States; The North Florida/South Georgia Veterans' Administration Medical Center, Gainesville, FL, 32610, United States
| |
Collapse
|
28
|
Animal models of excessive aggression: implications for human aggression and violence. Curr Opin Psychol 2017; 19:81-87. [PMID: 29279228 DOI: 10.1016/j.copsyc.2017.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/19/2022]
Abstract
Escalated interpersonal aggression and violence are common symptoms of multiple psychiatric disorders and represent a significant global health issue. Current therapeutic strategies are limited due to a lack of understanding about the neural and molecular mechanisms underlying the 'vicious' shift of normal adaptive aggression into violence, and the environmental triggers that cause it. Development of novel animal models that validly capture the salient features of human violent actions combined with newly emerging technologies for mapping, measuring, and manipulating neuronal activity in the brain significantly advance our understanding of the etiology, neuromolecular mechanisms, and potential therapeutic interventions of excessive aggressive behaviors in humans.
Collapse
|
29
|
Dobrzanski G, Kossut M. Application of the DREADD technique in biomedical brain research. Pharmacol Rep 2017; 69:213-221. [DOI: 10.1016/j.pharep.2016.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 12/25/2022]
|
30
|
Lo MC, Widge AS. Closed-loop neuromodulation systems: next-generation treatments for psychiatric illness. Int Rev Psychiatry 2017; 29:191-204. [PMID: 28523978 PMCID: PMC5461950 DOI: 10.1080/09540261.2017.1282438] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/10/2017] [Indexed: 01/19/2023]
Abstract
Despite deep brain stimulation's positive early results in psychiatric disorders, well-designed clinical trials have yielded inconsistent clinical outcomes. One path to more reliable benefit is closed-loop therapy: stimulation that is automatically adjusted by a device or algorithm in response to changes in the patient's electrical brain activity. These interventions may provide more precise and patient-specific treatments. This article first introduces the available closed-loop neuromodulation platforms, which have shown clinical efficacy in epilepsy and strong early results in movement disorders. It discusses the strengths and limitations of these devices in the context of psychiatric illness. It then describes emerging technologies to address these limitations, including pre-clinical developments such as wireless deep neurostimulation and genetically targeted neuromodulation. Finally, ongoing challenges and limitations for closed-loop psychiatric brain stimulation development, most notably the difficulty of identifying meaningful biomarkers for titration, are discussed. This is considered in the recently-released Research Domain Criteria (RDoC) framework, and how neuromodulation and RDoC are jointly very well suited to address the problem of treatment-resistant illness is described.
Collapse
Affiliation(s)
- Meng-Chen Lo
- Department of Psychiatry, Massachusetts General Hospital, Charlestown, MA
| | - Alik S. Widge
- Department of Psychiatry, Massachusetts General Hospital, Charlestown, MA
| |
Collapse
|
31
|
Layer-specific optogenetic activation of pyramidal neurons causes beta-gamma entrainment of neonatal networks. Nat Commun 2017; 8:14563. [PMID: 28216627 PMCID: PMC5321724 DOI: 10.1038/ncomms14563] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
Coordinated activity patterns in the developing brain may contribute to the wiring of neuronal circuits underlying future behavioural requirements. However, causal evidence for this hypothesis has been difficult to obtain owing to the absence of tools for selective manipulation of oscillations during early development. We established a protocol that combines optogenetics with electrophysiological recordings from neonatal mice in vivo to elucidate the substrate of early network oscillations in the prefrontal cortex. We show that light-induced activation of layer II/III pyramidal neurons that are transfected by in utero electroporation with a high-efficiency channelrhodopsin drives frequency-specific spiking and boosts network oscillations within beta–gamma frequency range. By contrast, activation of layer V/VI pyramidal neurons causes nonspecific network activation. Thus, entrainment of neonatal prefrontal networks in fast rhythms relies on the activation of layer II/III pyramidal neurons. This approach used here may be useful for further interrogation of developing circuits, and their behavioural readout. Oscillations in cortical activity during development are important for functional maturation. Here, the authors use optogenetics in neonatal mice to determine a causal role for pyramidal cell firing in different prelimbic cortex layers in generating beta–gamma range activity.
Collapse
|
32
|
Affiliation(s)
- Jeremy S Biane
- Department of Psychiatry and Center for Integrative Neuroscience, University of California, San Francisco, California, USA
| | - Mazen A Kheirbek
- Department of Psychiatry and Center for Integrative Neuroscience, University of California, San Francisco, California, USA,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California, USA,Weill Institute for Neurosciences, University of California, San Francisco, California, USA,E-mail:
| |
Collapse
|
33
|
Ciccocioppo R. Grand Challenge in Psychopharmacology: Setting Priorities to Shape a Bright Future. Front Psychiatry 2017; 8:15. [PMID: 28239360 PMCID: PMC5301004 DOI: 10.3389/fpsyt.2017.00015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/23/2017] [Indexed: 01/11/2023] Open
Affiliation(s)
- Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino , Camerino , Italy
| |
Collapse
|
34
|
Towne C, Thompson KR. Overview on Research and Clinical Applications of Optogenetics. ACTA ACUST UNITED AC 2016; 75:11.19.1-11.19.21. [DOI: 10.1002/cpph.13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Chris Towne
- Circuit Therapeutics, Inc Menlo Park California
| | | |
Collapse
|
35
|
Lenz M, Vlachos A. Releasing the Cortical Brake by Non-Invasive Electromagnetic Stimulation? rTMS Induces LTD of GABAergic Neurotransmission. Front Neural Circuits 2016; 10:96. [PMID: 27965542 PMCID: PMC5124712 DOI: 10.3389/fncir.2016.00096] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022] Open
Abstract
Repetitive Transcranial Magnetic Stimulation (rTMS) is a non-invasive brain stimulation technique which modulates cortical excitability beyond the stimulation period. However, despite its clinical use rTMS-based therapies which prevent or reduce disabilities in a functionally significant and sustained manner are scarce. It remains unclear how rTMS-mediated changes in cortical excitability, which are not task- or input-specific, exert beneficial effects in some healthy subjects and patients. While experimental evidence exists that repetitive magnetic stimulation (rMS) is linked to the induction of long-term potentiation (LTP) of excitatory neurotransmission, less attention has been dedicated to rTMS-induced structural, functional and molecular adaptations at inhibitory synapses. In this review article we provide a concise overview on basic neuroscience research, which reveals an important role of local disinhibitory networks in promoting associative learning and memory. These studies suggest that a reduction in inhibitory neurotransmission facilitates the expression of associative plasticity in cortical networks under physiological conditions. Hence, it is interesting to speculate that rTMS may act by decreasing GABAergic neurotransmission onto cortical principal neurons. Indeed, evidence has been provided that rTMS is capable of modulating inhibitory networks. Consistent with this suggestion recent basic science work discloses that a 10 Hz rTMS protocol reduces GABAergic synaptic strength on principal neurons. These findings support a model in which rTMS-induced long-term depression (LTD) of GABAergic synaptic strength mediates changes in excitation/inhibition-balance of cortical networks, which may in turn facilitate (or restore) the ability of stimulated networks to express input- and task-specific associative synaptic plasticity.
Collapse
Affiliation(s)
- Maximilian Lenz
- Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| | - Andreas Vlachos
- Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| |
Collapse
|
36
|
Copits BA, Pullen MY, Gereau RW. Spotlight on pain: optogenetic approaches for interrogating somatosensory circuits. Pain 2016; 157:2424-2433. [PMID: 27340912 PMCID: PMC5069102 DOI: 10.1097/j.pain.0000000000000620] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Bryan A Copits
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
37
|
Neuronal Signaling: an introduction. Neuronal Signal 2016; 1:NS20160025. [PMID: 32714575 PMCID: PMC7377261 DOI: 10.1042/ns20160025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
There have been a number of advances in our knowledge of neuronal communication in processes involved in development, functioning and disorders of the nervous system. This progress has prompted the Biochemical Society to launch Neuronal Signaling, a new open access journal that aims to expand on the existing knowledge about signaling within and between neurons.
Collapse
|
38
|
Abstract
Schizophrenia is a complex, heterogeneous behavioural and cognitive syndrome that seems to originate from disruption of brain development caused by genetic or environmental factors, or both. Dysfunction of dopaminergic neurotransmission contributes to the genesis of psychotic symptoms, but evidence also points to a widespread and variable involvement of other brain areas and circuits. Disturbances of synaptic function might underlie abnormalities of neuronal connectivity that possibly involves interneurons, but the precise nature, location, and timing of these events are uncertain. At present, treatment mainly consists of antipsychotic drugs combined with psychological therapies, social support, and rehabilitation, but a pressing need for more effective treatments and delivery of services exists. Advances in genomics, epidemiology, and neuroscience have led to great progress in understanding the disorder, and the opportunities for further scientific breakthrough are numerous--but so are the challenges.
Collapse
Affiliation(s)
- Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK.
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Preben B Mortensen
- Department of Economics, School of Business and Social Science, Aarhus University, Aarhus, Denmark
| |
Collapse
|
39
|
Abstract
Different cortical areas are organized into distinct intra-cortical subnetworks. How descending pathways from the entire cortex interact subcortically as a network remains unclear. Here, we report an open-access comprehensive mesoscale cortico-striatal projectome—a detailed connectivity projection map from the entire cerebral cortex to the dorsal striatum or caudoputamen (CP) in rodents. Based on these projections, we use novel computational neuroanatomical tools to identify 29 distinct functional striatal domains. Further, we characterize different cortico-striatal networks and how they reconfigure across the rostral-caudal extent of the CP. The workflow was also applied to select cortico-striatal connections in two different mouse models of disconnection syndromes to demonstrate its utility in characterizing circuitry-specific connectopathies. Together, this work provides the structural basis for studying the functional diversity of the dorsal striatum and disruptions of cortico-basal ganglia networks across a broad range of disorders.
Collapse
|
40
|
Saunders BT, Richard JM, Janak PH. Contemporary approaches to neural circuit manipulation and mapping: focus on reward and addiction. Philos Trans R Soc Lond B Biol Sci 2016; 370:20140210. [PMID: 26240425 DOI: 10.1098/rstb.2014.0210] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tying complex psychological processes to precisely defined neural circuits is a major goal of systems and behavioural neuroscience. This is critical for understanding adaptive behaviour, and also how neural systems are altered in states of psychopathology, such as addiction. Efforts to relate psychological processes relevant to addiction to activity within defined neural circuits have been complicated by neural heterogeneity. Recent advances in technology allow for manipulation and mapping of genetically and anatomically defined neurons, which when used in concert with sophisticated behavioural models, have the potential to provide great insight into neural circuit bases of behaviour. Here we discuss contemporary approaches for understanding reward and addiction, with a focus on midbrain dopamine and cortico-striato-pallidal circuits.
Collapse
Affiliation(s)
- Benjamin T Saunders
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jocelyn M Richard
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Patricia H Janak
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
41
|
Young CB, Chen T, Nusslock R, Keller J, Schatzberg AF, Menon V. Anhedonia and general distress show dissociable ventromedial prefrontal cortex connectivity in major depressive disorder. Transl Psychiatry 2016; 6:e810. [PMID: 27187232 PMCID: PMC5070048 DOI: 10.1038/tp.2016.80] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 03/27/2016] [Accepted: 03/31/2016] [Indexed: 12/18/2022] Open
Abstract
Anhedonia, the reduced ability to experience pleasure in response to otherwise rewarding stimuli, is a core symptom of major depressive disorder (MDD). Although the posterior ventromedial prefrontal cortex (pVMPFC) and its functional connections have been consistently implicated in MDD, their roles in anhedonia remain poorly understood. Furthermore, it is unknown whether anhedonia is primarily associated with intrinsic 'resting-state' pVMPFC functional connectivity or an inability to modulate connectivity in a context-specific manner. To address these gaps, a pVMPFC region of interest was first identified using activation likelihood estimation meta-analysis. pVMPFC connectivity was then examined in relation to anhedonia and general distress symptoms of depression, using both resting-state and task-based functional magnetic resonance imaging involving pleasant music, in current MDD and healthy control groups. In MDD, pVMPFC connectivity was negatively correlated with anhedonia but not general distress during music listening in key reward- and emotion-processing regions, including nucleus accumbens, ventral tegmental area/substantia nigra, orbitofrontal cortex and insula, as well as fronto-temporal regions involved in tracking complex sound sequences, including middle temporal gyrus and inferior frontal gyrus. No such dissociations were observed in the healthy controls, and resting-state pVMPFC connectivity did not dissociate anhedonia from general distress in either group. Our findings demonstrate that anhedonia in MDD is associated with context-specific deficits in pVMPFC connectivity with the mesolimbic reward system when encountering pleasurable stimuli, rather than a static deficit in intrinsic resting-state connectivity. Critically, identification of functional circuits associated with anhedonia better characterizes MDD heterogeneity and may help track of one of its core symptoms.
Collapse
Affiliation(s)
- C B Young
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Psychology, Northwestern University, Evanston, IL, USA
| | - T Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - R Nusslock
- Department of Psychology, Northwestern University, Evanston, IL, USA
| | - J Keller
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - A F Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - V Menon
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
42
|
Quach TT, Lerch JK, Honnorat J, Khanna R, Duchemin AM. Neuronal networks in mental diseases and neuropathic pain: Beyond brain derived neurotrophic factor and collapsin response mediator proteins. World J Psychiatry 2016; 6:18-30. [PMID: 27014595 PMCID: PMC4804265 DOI: 10.5498/wjp.v6.i1.18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/24/2015] [Accepted: 01/07/2016] [Indexed: 02/05/2023] Open
Abstract
The brain is a complex network system that has the capacity to support emotion, thought, action, learning and memory, and is characterized by constant activity, constant structural remodeling, and constant attempt to compensate for this remodeling. The basic insight that emerges from complex network organization is that substantively different networks can share common key organizational principles. Moreover, the interdependence of network organization and behavior has been successfully demonstrated for several specific tasks. From this viewpoint, increasing experimental/clinical observations suggest that mental disorders are neural network disorders. On one hand, single psychiatric disorders arise from multiple, multifactorial molecular and cellular structural/functional alterations spreading throughout local/global circuits leading to multifaceted and heterogeneous clinical symptoms. On the other hand, various mental diseases may share functional deficits across the same neural circuit as reflected in the overlap of symptoms throughout clinical diagnoses. An integrated framework including experimental measures and clinical observations will be necessary to formulate a coherent and comprehensive understanding of how neural connectivity mediates and constraints the phenotypic expression of psychiatric disorders.
Collapse
|
43
|
Lerner TN, Ye L, Deisseroth K. Communication in Neural Circuits: Tools, Opportunities, and Challenges. Cell 2016; 164:1136-1150. [PMID: 26967281 PMCID: PMC5725393 DOI: 10.1016/j.cell.2016.02.027] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/27/2016] [Accepted: 02/03/2016] [Indexed: 11/27/2022]
Abstract
Communication, the effective delivery of information, is fundamental to life across all scales and species. Nervous systems (by necessity) may be most specifically adapted among biological tissues for high rate and complexity of information transmitted, and thus, the properties of neural tissue and principles of its organization into circuits may illuminate capabilities and limitations of biological communication. Here, we consider recent developments in tools for studying neural circuits with particular attention to defining neuronal cell types by input and output information streams--i.e., by how they communicate. Complementing approaches that define cell types by virtue of genetic promoter/enhancer properties, this communication-based approach to defining cell types operationally by input/output (I/O) relationships links structure and function, resolves difficulties associated with single-genetic-feature definitions, leverages technology for observing and testing significance of precisely these I/O relationships in intact brains, and maps onto processes through which behavior may be adapted during development, experience, and evolution.
Collapse
Affiliation(s)
- Talia N Lerner
- Bioengineering Department, 318 Campus Drive, Stanford University, Stanford, CA 94305, USA
| | - Li Ye
- Bioengineering Department, 318 Campus Drive, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Bioengineering Department, 318 Campus Drive, Stanford University, Stanford, CA 94305, USA; Psychiatry Department, 318 Campus Drive, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, 318 Campus Drive, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
44
|
Animal models of major depression and their clinical implications. Prog Neuropsychopharmacol Biol Psychiatry 2016; 64:293-310. [PMID: 25891248 DOI: 10.1016/j.pnpbp.2015.04.004] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/09/2015] [Accepted: 04/12/2015] [Indexed: 12/12/2022]
Abstract
Major depressive disorder is a common, complex, and potentially life-threatening mental disorder that imposes a severe social and economic burden worldwide. Over the years, numerous animal models have been established to elucidate pathophysiology that underlies depression and to test novel antidepressant treatment strategies. Despite these substantial efforts, the animal models available currently are of limited utility for these purposes, probably because none of the models mimics this complex disorder fully. It is presumable that psychiatric illnesses, such as affective disorders, are related to the complexity of the human brain. Here, we summarize the animal models that are used most commonly for depression, and discuss their advantages and limitations. We discuss genetic models, including the recently developed optogenetic tools and the stress models, such as the social stress, chronic mild stress, learned helplessness, and early-life stress paradigms. Moreover, we summarize briefly the olfactory bulbectomy model, as well as models that are based on pharmacological manipulations and disruption of the circadian rhythm. Finally, we highlight common misinterpretations and often-neglected important issues in this field.
Collapse
|
45
|
Caution When Diagnosing Your Mouse With Schizophrenia: The Use and Misuse of Model Animals for Understanding Psychiatric Disorders. Biol Psychiatry 2016; 79:32-8. [PMID: 26058706 DOI: 10.1016/j.biopsych.2015.04.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/26/2015] [Accepted: 04/23/2015] [Indexed: 12/14/2022]
Abstract
Animal models are widely used in biomedical research, but their applicability to psychiatric disorders is less clear. There are several reasons for this, including 1) emergent features of psychiatric illness that are not captured by the sum of individual symptoms, 2) a lack of equivalency between model animal behavior and human psychiatric symptoms, and 3) the possibility that model organisms do not have (and may not be capable of having) the same illnesses as humans. Here, we discuss the effective use, and inherent limitations, of model animals for psychiatric research. As disrupted-in-schizophrenia 1 (DISC1) is a genetic risk factor across a spectrum of psychiatric disorders, we focus on the results of studies using mice with various mutations of DISC1. The data from a broad range of studies show remarkable consistency with the effects of DISC1 mutation on developmental/anatomical endophenotypes. However, when one expands the phenotype to include behavioral correlates of human psychiatric diseases, much of this consistency ends. Despite these challenges, model animals remain valuable for understanding the basic brain processes that underlie psychiatric diseases. We argue that model animals have great potential to help us understand the core neurobiological dysfunction underlying psychiatric disorders and that marrying genetics and brain circuits with behavior is a good way forward.
Collapse
|
46
|
Of Mice, Men, and Microbial Opsins: How Optogenetics Can Help Hone Mouse Models of Mental Illness. Biol Psychiatry 2016; 79:47-52. [PMID: 25981174 PMCID: PMC4618781 DOI: 10.1016/j.biopsych.2015.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 01/04/2023]
Abstract
Genetic, pharmacologic, and behavioral manipulations have long been powerful tools for generating rodent models to study the neural substrates underlying psychiatric disease. Recent advances in the use of optogenetics in awake behaving rodents has added an additional valuable methodology to this experimental toolkit. Here, we review several recent studies that leverage optogenetic technologies to elucidate neural mechanisms possibly related to depression, anxiety, and obsessive-compulsive disorder. We use a few illustrative examples to highlight key emergent principles about how optogenetics, in conjunction with more established modalities, can help to organize our understanding of how disease-related states, specific neuronal circuits, and various behavioral assays fit into hierarchical frameworks such as the National Institute of Mental Health Research Domain Criteria matrix.
Collapse
|
47
|
Fuccillo MV, Rothwell PE, Malenka RC. From Synapses to Behavior: What Rodent Models Can Tell Us About Neuropsychiatric Disease. Biol Psychiatry 2016; 79:4-6. [PMID: 26616432 PMCID: PMC4832414 DOI: 10.1016/j.biopsych.2015.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/11/2015] [Indexed: 10/22/2022]
|
48
|
Song C, Knöpfel T. Optogenetics enlightens neuroscience drug discovery. Nat Rev Drug Discov 2015; 15:97-109. [DOI: 10.1038/nrd.2015.15] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
49
|
Abstract
Anhedonia, or the loss of pleasure in previously rewarding stimuli, is a core symptom of major depressive disorder that may reflect an underlying dysregulation in reward processing. The mesolimbic dopamine circuit, also known as the brain's reward circuit, is integral to processing the rewarding salience of stimuli to guide actions. Manifestation of anhedonia and associated depression symptoms like feelings of sadness, changes in appetite, and psychomotor effects, may reflect changes in the brain reward circuitry as a common underlying disease process. This review will synthesize the recent literature from human and rodent studies providing a circuit-level framework for understanding anhedonia in depression, with emphasis on the nucleus accumbens.
Collapse
Affiliation(s)
- Mitra Heshmati
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Icahn 10-71, Box 1065, New York, NY 10029 (212) 659- 5917
| | - Scott J Russo
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Icahn 10-71, Box 1065, New York, NY 10029 (212) 659- 5917
| |
Collapse
|
50
|
Heckenast JR, Wilkinson LS, Jones MW. Decoding Advances in Psychiatric Genetics: A Focus on Neural Circuits in Rodent Models. ADVANCES IN GENETICS 2015; 92:75-106. [PMID: 26639916 DOI: 10.1016/bs.adgen.2015.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Appropriately powered genome-wide association studies combined with deep-sequencing technologies offer the prospect of real progress in revealing the complex biological underpinnings of schizophrenia and other psychiatric disorders. Meanwhile, recent developments in genome engineering, including CRISPR, constitute better tools to move forward with investigating these genetic leads. This review aims to assess how these advances can inform the development of animal models for psychiatric disease, with a focus on schizophrenia and in vivo electrophysiological circuit-level measures with high potential as disease biomarkers.
Collapse
Affiliation(s)
- Julia R Heckenast
- School of Psychology, Cardiff University, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK; Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Lawrence S Wilkinson
- School of Psychology, Cardiff University, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK; Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Matthew W Jones
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, UK
| |
Collapse
|