1
|
Jha NA, Ayoub SM, Flesher MM, Morton K, Sikkink M, de Guglielmo G, Khokhar JY, Minassian A, Brody AL, Young JW. Acute nicotine vapor normalizes sensorimotor gating and reduces locomotor activity deficits in HIV-1 transgenic rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599641. [PMID: 38948796 PMCID: PMC11212989 DOI: 10.1101/2024.06.18.599641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Rationale Despite improved life expectancy of people with HIV (PWH), HIV-associated neurocognitive impairment (NCI) persists, alongside deficits in sensorimotor gating and neuroinflammation. PWH exhibit high smoking rates, possibly due to neuroprotective, anti-inflammatory, and cognitive-enhancing effects of nicotine, suggesting potential self-medication. Objectives Here, we tested the effects of acute nicotine vapor exposure on translatable measures of sensorimotor gating and exploratory behavior in the HIV-1 transgenic (HIV-1Tg) rat model of HIV. Methods Male and female HIV-1Tg and F344 control rats (n=57) were exposed to acute nicotine or vehicle vapor. Sensorimotor gating was assessed using prepulse inhibition (PPI) of the acoustic startle response, and exploratory behavior was evaluated using the behavioral pattern monitor (BPM). Results Vehicle-treated HIV-1Tg rats exhibited PPI deficits at low prepulse intensities compared to F344 controls, as seen previously. No PPI deficits were observed in nicotine-treated HIV1-Tg rats, however. HIV-1Tg rats were hypoactive in the BPM relative to controls, whilst nicotine vapor increased activity and exploratory behavior across genotypes. Cotinine analyses confirmed comparable levels of the primary metabolite of nicotine across genotypes. Conclusions Previous findings of PPI deficits in HIV-1Tg rats were replicated and, importantly, attenuated by acute nicotine vapor. Evidence for similar cotinine levels suggest a nicotine-specific effect in HIV-1Tg rats. HIV-1Tg rats had reduced exploratory behavior compared to controls, attenuated by acute nicotine vapor. Therefore, acute nicotine may be beneficial for remediating sensorimotor and locomotor activity deficits in PWH. Future studies should determine the long-term effects of nicotine vapor on similar HIV/NCI-relevant behaviors.
Collapse
Affiliation(s)
- Neal A. Jha
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Samantha M. Ayoub
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - M. Melissa Flesher
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kathleen Morton
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Megan Sikkink
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Giordano de Guglielmo
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jibran Y. Khokhar
- Department of Anatomy and Cell Biology, University of Western Ontario 1151 Richmond Street, London, ON N61 3K7, Canada
| | - Arpi Minassian
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
- Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| | - Arthur L. Brody
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
- Research Service, VA San Diego Healthcare System 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| | - Jared W. Young
- Department of Psychiatry, University of California, San Diego 9500 Gilman Drive, La Jolla, CA 92093, USA
- Research Service, VA San Diego Healthcare System 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| |
Collapse
|
2
|
Chaudhary R, Singh R. Therapeutic Viewpoint on Rat Models of Locomotion Abnormalities and Neurobiological Indicators in Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:488-503. [PMID: 37202886 DOI: 10.2174/1871527322666230518111323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/11/2022] [Accepted: 12/02/2022] [Indexed: 05/20/2023]
Abstract
BACKGROUND Locomotion problems in Parkinson's syndrome are still a research and treatment difficulty. With the recent introduction of brain stimulation or neuromodulation equipment that is sufficient to monitor activity in the brain using electrodes placed on the scalp, new locomotion investigations in patients having the capacity to move freely have sprung up. OBJECTIVE This study aimed to find rat models and locomotion-connected neuronal indicators and use them all over a closed-loop system to enhance the future and present treatment options available for Parkinson's disease. METHODS Various publications on locomotor abnormalities, Parkinson's disease, animal models, and other topics have been searched using several search engines, such as Google Scholar, Web of Science, Research Gate, and PubMed. RESULTS Based on the literature, we can conclude that animal models are used for further investigating the locomotion connectivity deficiencies of many biological measuring devices and attempting to address unanswered concerns from clinical and non-clinical research. However, translational validity is required for rat models to contribute to the improvement of upcoming neurostimulation-based medicines. This review discusses the most successful methods for modelling Parkinson's locomotion in rats. CONCLUSION This review article has examined how scientific clinical experiments lead to localised central nervous system injuries in rats, as well as how the associated motor deficits and connection oscillations reflect this. This evolutionary process of therapeutic interventions may help to improve locomotion- based treatment and management of Parkinson's syndrome in the upcoming years.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
- Department of Pharmacology, M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
| |
Collapse
|
3
|
Markam PS, Bourguignon C, Zhu L, Darvas M, Sabatini PV, Kokoeva MV, Giros B, Storch KF. The neurons that drive infradian sleep-wake and mania-like behavioral rhythms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.566955. [PMID: 38014299 PMCID: PMC10680706 DOI: 10.1101/2023.11.14.566955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Infradian mood and sleep-wake rhythms with periods of 48 hr and beyond have been observed in bipolar disorder (BD) subjects that even persist in time isolation, indicating an endogenous origin. Here we show that mice exposed to methamphetamine (Meth) in drinking water develop infradian locomotor rhythms with periods of 48 hr and beyond which extend to sleep length and mania-like behaviors in support of a model for cycling in BD. This cycling capacity is abrogated upon genetic disruption of DA production in DA neurons of the ventral tegmental area (VTA) or ablation of nucleus accumbens (NAc) projecting, dopamine (DA) neurons. Chemogenetic activation of NAc-projecting DA neurons leads to locomotor period lengthening in clock deficient mice, while cytosolic calcium in DA processes of the NAc was found fluctuating synchronously with locomotor behavior. Together, our findings argue that BD cycling relies on infradian rhythm generation that depends on NAc-projecting DA neurons.
Collapse
|
4
|
Young JW. Development of cross-species translational paradigms for psychiatric research in the Research Domain Criteria era. Neurosci Biobehav Rev 2023; 148:105119. [PMID: 36889561 DOI: 10.1016/j.neubiorev.2023.105119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
The past 30 years of IBNS has included research attempting to treat the cognitive and behavioral deficits observed in people with psychiatric conditions. Early work utilized drugs identified from tests thought to be cognition-relevant, however the high failure rate crossing the translational-species barrier led to focus on developing valid cross-species translational tests. The face, predictive, and neurobiological validities used to assess animal models of psychiatry can be used to validate these tests. Clinical sensitivity is another important aspect however, for if the clinical population targeted for treatment does not exhibit task deficits, then why develop treatments? This review covers some work validating cross-species translational tests and suggests future directions. Also covered is the contribution IBNS made to fostering such research and my role in IBNS, making it more available to all including fostering mentor/mentee programs plus spearheading diversity and inclusivity initiatives. All science needs support and IBNS has supported research recreating the behavioral abnormalities that define psychiatric conditions with the aim to improve the lives of people with such conditions.
Collapse
Affiliation(s)
- Jared W Young
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
5
|
Canzian J, Gonçalves FLS, Müller TE, Franscescon F, Santos LW, Adedara IA, Rosemberg DB. Zebrafish as a potential non-traditional model organism in translational bipolar disorder research: Genetic and behavioral insights. Neurosci Biobehav Rev 2022; 136:104620. [PMID: 35300991 DOI: 10.1016/j.neubiorev.2022.104620] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/16/2022] [Accepted: 03/10/2022] [Indexed: 01/14/2023]
Abstract
Bipolar disorder (BD) is a severe and debilitating illness that affects 1-2% of the population worldwide. BD is characterized by recurrent and extreme mood swings, including mania/hypomania and depression. Animal experimental models have been used to elucidate the mechanisms underlying BD and different strategies have been proposed to assess BD-like symptoms. The zebrafish (Danio rerio) has been considered a suitable vertebrate system for modeling BD-like responses, due to the genetic tractability, molecular/physiological conservation, and well-characterized behavioral responses. In this review, we discuss how zebrafish-based models can be successfully used to understand molecular, biochemical, and behavioral alterations paralleling those found in BD. We also outline some advantages and limitations of this aquatic species to examine BD-like phenotypes in translational neurobehavioral research. Overall, we reinforce the use of zebrafish as a promising tool to investigate the neural basis associated with BD-like behaviors, which may foster the discovery of novel pharmacological therapies.
Collapse
Affiliation(s)
- Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Falco L S Gonçalves
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Talise E Müller
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Francini Franscescon
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Laura W Santos
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Isaac A Adedara
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
6
|
Bohnen NI, Costa RM, Dauer WT, Factor SA, Giladi N, Hallett M, Lewis SJ, Nieuwboer A, Nutt JG, Takakusaki K, Kang UJ, Przedborski S, Papa SM. Discussion of Research Priorities for Gait Disorders in Parkinson's Disease. Mov Disord 2021; 37:253-263. [PMID: 34939221 PMCID: PMC10122497 DOI: 10.1002/mds.28883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/08/2021] [Accepted: 11/10/2021] [Indexed: 12/18/2022] Open
Abstract
Gait and balance abnormalities develop commonly in Parkinson's disease and are among the motor symptoms most disabling and refractory to dopaminergic or other treatments, including deep brain stimulation. Efforts to develop effective therapies are challenged by limited understanding of these complex disorders. There is a major need for novel and appropriately targeted research to expedite progress in this area. The Scientific Issues Committee of the International Parkinson and Movement Disorder Society has charged a panel of experts in the field to consider the current knowledge gaps and determine the research routes with highest potential to generate groundbreaking data. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Nicolaas I. Bohnen
- Departments of Radiology and Neurology University of Michigan and VA Ann Arbor Healthcare System Ann Arbor Michigan USA
| | - Rui M. Costa
- Departments of Neuroscience and Neurology, Zuckerman Mind Brain Behavior Institute Columbia University New York New York USA
| | - William T. Dauer
- Departments of Neurology and Neuroscience The Peter O'Donnell Jr. Brain Institute, UT Southwestern Dallas Texas USA
| | - Stewart A. Factor
- Jean and Paul Amos Parkinson's Disease and Movement Disorders Program Emory University School of Medicine Atlanta Georgia USA
| | - Nir Giladi
- Movement Disorders Unit, Department of Neurology, Tel‐Aviv Sourasky Medical Center, Sackler School of Medicine and Sagol School of Neuroscience Tel Aviv University Tel Aviv Israel
| | - Mark Hallett
- Human Motor Control Section National Institute of Neurological Disorders and Stroke, National Institutes of Health Bethesda Maryland USA
| | - Simon J.G. Lewis
- ForeFront Parkinson's Disease Research Clinic, Brain and Mind Centre, School of Medical Sciences University of Sydney Sydney New South Wales Australia
| | - Alice Nieuwboer
- Department of Rehabilitation Sciences KU Leuven Leuven Belgium
| | - John G. Nutt
- Movement Disorder Section, Department of Neurology Oregon Health & Science University Portland Oregon USA
| | - Kaoru Takakusaki
- Department of Physiology, Section of Neuroscience Asahikawa Medical University Asahikawa Japan
| | - Un Jung Kang
- Departments of Neurology, Neuroscience, and Physiology Neuroscience Institute, The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, The Parekh Center for Interdisciplinary Neurology, New York University Grossman School of Medicine New York New York USA
| | - Serge Przedborski
- Departments of Pathology and Cell Biology, Neurology, and Neuroscience Columbia University New York New York USA
| | - Stella M. Papa
- Department of Neurology, School of Medicine, and Yerkes National Primate Research Center Emory University Atlanta Georgia USA
| | | |
Collapse
|
7
|
Enlightened: addressing circadian and seasonal changes in photoperiod in animal models of bipolar disorder. Transl Psychiatry 2021; 11:373. [PMID: 34226504 PMCID: PMC8257630 DOI: 10.1038/s41398-021-01494-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
Bipolar disorders (BDs) exhibit high heritability and symptoms typically first occur during late adolescence or early adulthood. Affected individuals may experience alternating bouts of mania/hypomania and depression, with euthymic periods of varying lengths interspersed between these extremes of mood. Clinical research studies have consistently demonstrated that BD patients have disturbances in circadian and seasonal rhythms, even when they are free of symptoms. In addition, some BD patients display seasonal patterns in the occurrence of manic/hypomanic and depressive episodes as well as the time of year when symptoms initially occur. Finally, the age of onset of BD symptoms is strongly influenced by the distance one lives from the equator. With few exceptions, animal models useful in the study of BD have not capitalized on these clinical findings regarding seasonal patterns in BD to explore molecular mechanisms associated with the expression of mania- and depression-like behaviors in laboratory animals. In particular, animal models would be especially useful in studying how rates of change in photoperiod that occur during early spring and fall interact with risk genes to increase the occurrence of mania- and depression-like phenotypes, respectively. Another unanswered question relates to the ways in which seasonally relevant changes in photoperiod affect responses to acute and chronic stressors in animal models. Going forward, we suggest ways in which translational research with animal models of BD could be strengthened through carefully controlled manipulations of photoperiod to enhance our understanding of mechanisms underlying seasonal patterns of BD symptoms in humans. In addition, we emphasize the value of incorporating diurnal rodent species as more appropriate animal models to study the effects of seasonal changes in light on symptoms of depression and mania that are characteristic of BD in humans.
Collapse
|
8
|
Kwiatkowski MA, Roberts BZ, van Enkhuizen J, Ji B, Zhou X, Young JW. Chronic nicotine, but not suramin or resveratrol, partially remediates the mania-like profile of dopamine transporter knockdown mice. Eur Neuropsychopharmacol 2021; 42:75-86. [PMID: 33191077 PMCID: PMC8853461 DOI: 10.1016/j.euroneuro.2020.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 10/19/2020] [Accepted: 11/02/2020] [Indexed: 01/17/2023]
Abstract
Bipolar disorder (BD) is a severe mental illness affecting 2% of the global population. Current pharmacotherapies provide incomplete symptom remediation, highlighting the need for novel therapeutics. BD is characterized by fluctuations between mania and depression, likely driven by shifts between hyperdopaminergia and hypercholinergia, respectively. Hyperdopaminergia may result from insufficient activity of the dopamine transporter (DAT), the primary mediator of synaptic dopamine clearance. The DAT knockdown (DAT KD) mouse recreates this mechanism and exhibits a highly reproducible hyperexploratory profile in the cross-species translatable Behavioral Pattern Monitor (BPM) that is: (a) consistent with that observed in BD mania patients; and (b) partially normalized by chronic lithium and valproate treatment. The DAT KD/BPM model of mania therefore exhibits high levels of face-, construct-, and predictive-validity for the pre-clinical assessment of putative anti-mania drugs. Three different drug regimens - chronic nicotine (nicotinic acetylcholine receptor (nAChR) agonist; 40 mg/kg/d, 26 d), subchronic suramin (anti-purinergic; 20 mg/kg, 1 × /wk, 4 wks), and subchronic resveratrol (striatal DAT upregulator; 20 mg/kg/d, 4 d) - were administered to separate cohorts of male and female DAT KD- and wildtype (WT) littermate mice, and exploration was assessed in the BPM. Throughout, DAT KD mice exhibited robust hyperexploratory profiles relative to WTs. Nicotine partially normalized this behavior. Resveratrol modestly upregulated DAT expression but did not normalize DAT KD behavior. These results support the mania-like profile of DAT KD mice, which may be partially remediated by nAChR agonists via restoration of disrupted catecholaminergic/cholinergic equilibrium. Delineating the precise mechanism of action of nicotine could identify more selective therapeutic targets.
Collapse
Affiliation(s)
- Molly A Kwiatkowski
- Department of Psychiatry, UC San Diego School of Medicine, University of California, San Diego, CA 92093-0804, United States
| | - Benjamin Z Roberts
- Department of Psychiatry, UC San Diego School of Medicine, University of California, San Diego, CA 92093-0804, United States
| | - Jordy van Enkhuizen
- Department of Psychiatry, UC San Diego School of Medicine, University of California, San Diego, CA 92093-0804, United States
| | - Baohu Ji
- Department of Psychiatry, UC San Diego School of Medicine, University of California, San Diego, CA 92093-0804, United States
| | - Xianjin Zhou
- Department of Psychiatry, UC San Diego School of Medicine, University of California, San Diego, CA 92093-0804, United States
| | - Jared W Young
- Department of Psychiatry, UC San Diego School of Medicine, University of California, San Diego, CA 92093-0804, United States; Research Service, VA San Diego Healthcare System, United States.
| |
Collapse
|
9
|
Honda T, Takata Y, Cherasse Y, Mizuno S, Sugiyama F, Takahashi S, Funato H, Yanagisawa M, Lazarus M, Oishi Y. Ablation of Ventral Midbrain/Pons GABA Neurons Induces Mania-like Behaviors with Altered Sleep Homeostasis and Dopamine D 2R-mediated Sleep Reduction. iScience 2020; 23:101240. [PMID: 32563157 PMCID: PMC7305386 DOI: 10.1016/j.isci.2020.101240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/04/2020] [Accepted: 06/02/2020] [Indexed: 11/12/2022] Open
Abstract
Individuals with the neuropsychiatric disorder mania exhibit hyperactivity, elevated mood, and a decreased need for sleep. The brain areas and neuronal populations involved in mania-like behaviors, however, have not been elucidated. In this study, we found that ablating the ventral medial midbrain/pons (VMP) GABAergic neurons induced mania-like behaviors in mice, including hyperactivity, anti-depressive behaviors, reduced anxiety, increased risk-taking behaviors, distractibility, and an extremely shortened sleep time. Strikingly, these mice also showed no rebound sleep after sleep deprivation, suggesting abnormal sleep homeostatic regulation. Dopamine D2 receptor deficiency largely abolished the sleep reduction induced by ablating the VMP GABAergic neurons without affecting the hyperactivity and anti-depressive behaviors. Our data demonstrate that VMP GABAergic neurons are involved in the expression of mania-like behaviors, which can be segregated to the short-sleep and other phenotypes on the basis of the dopamine D2 receptors. Hyperactivity and anti-depressive behaviors are induced by loss of VMP GABA neurons Homeostatic sleep rebound is lost together with largely shorten daily sleep Dopamine D2 receptors mediate the daytime sleep loss
Collapse
Affiliation(s)
- Takato Honda
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA.
| | - Yohko Takata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Trans-border Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center and Trans-border Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Laboratory Animal Resource Center and Trans-border Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Anatomy, Faculty of Medicine, Toho University, Ota, Tokyo 143-8540, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Life Science Center for Survival Dynamics (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; R&D Center for Frontiers of Mirai in Policy and Technology (F-MIRAI), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
10
|
Kwiatkowski MA, Cope ZA, Lavadia ML, van de Cappelle CJA, Dulcis D, Young JW. Short-active photoperiod gestation induces psychiatry-relevant behavior in healthy mice but a resiliency to such effects are seen in mice with reduced dopamine transporter expression. Sci Rep 2020; 10:10217. [PMID: 32576854 PMCID: PMC7311429 DOI: 10.1038/s41598-020-66873-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/13/2020] [Indexed: 01/02/2023] Open
Abstract
A higher incidence of multiple psychiatric disorders occurs in people born in late winter/early spring. Reduced light exposure/activity level impacts adult rodent behavior and neural mechanisms, yet few studies have investigated such light exposure on gestating fetuses. A dysfunctional dopamine system is implicated in most psychiatric disorders, and genetic polymorphisms reducing expression of the dopamine transporter (DAT) are associated with some conditions. Furthermore, adult mice with reduced DAT expression (DAT-HT) were hypersensitive to short active (SA; 19:5 L:D) photoperiod exposure versus their wildtype (WT) littermates. Effects of SA photoperiod exposure during gestation in these mice have not been examined. We confirmed adult females exhibit a heightened corticosterone response when in SA photoperiod. We then tested DAT-HT mice and WT littermates in psychiatry-relevant behavioral tests after SA or normal active (NA; 12:12 L:D) photoperiod exposure during gestation and early life. SA-born WT mice exhibited sensorimotor gating deficits (males), increased reward preference, less immobility, open arm avoidance (females), less motivation to obtain a reward, and reversal learning deficits, vs. NA-born WT mice. DAT-HT mice were largely resilient to these effects, however. Future studies will determine the mechanism(s) by which SA photoperiod exposure influences brain development to predispose toward emergence of psychiatry-relevant behaviors.
Collapse
Affiliation(s)
- Molly A Kwiatkowski
- Department of Psychiatry, University of California, San Diego, San Diego, USA
| | - Zackary A Cope
- Department of Medicine, Aging Institute, University of Pittsburgh, Pittsburgh, USA
| | - Maria L Lavadia
- Department of Psychiatry, University of California, San Diego, San Diego, USA
| | - Chuck J A van de Cappelle
- Department of Psychiatry, University of California, San Diego, San Diego, USA.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Davide Dulcis
- Department of Psychiatry, University of California, San Diego, San Diego, USA
| | - Jared W Young
- Department of Psychiatry, University of California, San Diego, San Diego, USA. .,Research Service, VA San Diego Healthcare System, San Diego, USA.
| |
Collapse
|
11
|
Fulcher N, Azzopardi E, De Oliveira C, Hudson R, Schormans AL, Zaman T, Allman BL, Laviolette SR, Schmid S. Deciphering midbrain mechanisms underlying prepulse inhibition of startle. Prog Neurobiol 2019; 185:101734. [PMID: 31863802 DOI: 10.1016/j.pneurobio.2019.101734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/19/2019] [Accepted: 12/11/2019] [Indexed: 12/16/2022]
Abstract
Prepulse inhibition (PPI) is an operational measure of sensorimotor gating. Deficits of PPI are a hallmark of schizophrenia and associated with several other psychiatric illnesses such as e.g. autism spectrum disorder, yet the mechanisms underlying PPI are still not fully understood. There is growing evidence contradicting the long-standing hypothesis that PPI is mediated by a short feed-forward midbrain circuitry including inhibitory cholinergic projections from the pedunculopontine tegmental nucleus (PPTg) to the startle pathway. Here, we employed a chemogenetic approach to explore the involvement of the PPTg in general, and cholinergic neurons specifically, in PPI. Activation of inhibitory DREADDs (designer receptors exclusively activated by designer drugs) in the PPTg by systemic administration of clozapine-N-oxide (CNO) disrupted PPI, confirming the involvement of the PPTg in PPI. In contrast, chemogenetic inhibition of specifically cholinergic PPTg neurons had no effect on PPI, but inhibited morphine-induced conditioned place preference (CPP) in the same animals, showing that the DREADDs were effective in modulating behavior. These findings support a functional role of the PPTg and/or neighboring structures in PPI in accordance with previous lesion studies, but also provide strong evidence against the hypothesis that specifically cholinergic PPTg neurons are involved in mediating PPI, implicating rather non-cholinergic midbrain neurons.
Collapse
Affiliation(s)
- Niveen Fulcher
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada
| | - Erin Azzopardi
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Cleusa De Oliveira
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Roger Hudson
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada
| | - Ashley L Schormans
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Tariq Zaman
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Brian L Allman
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Steven R Laviolette
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Susanne Schmid
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada.
| |
Collapse
|
12
|
Krug JT, Klein AK, Purvis EM, Ayala K, Mayes MS, Collins L, Fisher MP, Ettenberg A. Effects of chronic lithium exposure in a modified rodent ketamine-induced hyperactivity model of mania. Pharmacol Biochem Behav 2019; 179:150-155. [DOI: 10.1016/j.pbb.2019.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 10/27/2022]
|
13
|
Kirlic N, Aupperle RL, Rhudy JL, Misaki M, Kuplicki R, Sutton A, Alvarez RP. Latent variable analysis of negative affect and its contributions to neural responses during shock anticipation. Neuropsychopharmacology 2019; 44:695-702. [PMID: 30181595 PMCID: PMC6372706 DOI: 10.1038/s41386-018-0187-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/11/2018] [Accepted: 08/14/2018] [Indexed: 01/04/2023]
Abstract
Negative affect is considered an important factor in the etiology of depression and anxiety, and is highly related to pain. However, negative affect is not a unitary construct. To identify specific targets for treatment development, we aimed to derive latent variables of negative affect and test their unique contributions to affective processing during anticipation of unpredictable, painful shock. Eighty-three subjects (43 with depression and anxiety spectrum disorders and 40 healthy controls) completed self-report measures of negative valence and underwent neuroimaging while exploring computer-simulated contexts with and without the threat of a painful, but tolerable, shock. Principal component analysis (PCA) extracted distinct components of general negative affect (GNA) and pain-related negative affect (PNA). While elevated GNA and PNA were both indicative of depression and anxiety disorders, greater PNA was more strongly related to task-specific anxious reactivity during shock anticipation. GNA was associated with increased precuneus and middle frontal gyrus activity, whereas PNA was related to increased bilateral anterior insula activity. Anterior insula activity mediated the relationship between PNA and task-specific anxious reactivity. In conclusion, GNA and PNA have distinct neural signatures and uniquely contribute to anxious anticipation. PNA, via insula activity, may relate to arousal in ways that could contribute to affective dysregulation, and thus may be an important treatment target.
Collapse
Affiliation(s)
- Namik Kirlic
- Laureate Institute for Brain Research, Tulsa, OK, USA.
| | - Robin L. Aupperle
- 0000 0004 0512 8863grid.417423.7Laureate Institute for Brain Research, Tulsa, OK USA ,0000 0001 2160 264Xgrid.267360.6School of Community Medicine, University of Tulsa, Tulsa, OK USA
| | - Jamie L. Rhudy
- 0000 0001 2160 264Xgrid.267360.6Department of Psychology, University of Tulsa, Tulsa, OK USA
| | - Masaya Misaki
- 0000 0004 0512 8863grid.417423.7Laureate Institute for Brain Research, Tulsa, OK USA
| | - Rayus Kuplicki
- 0000 0004 0512 8863grid.417423.7Laureate Institute for Brain Research, Tulsa, OK USA
| | - Anne Sutton
- 0000 0004 0512 8863grid.417423.7Laureate Institute for Brain Research, Tulsa, OK USA
| | - Ruben P. Alvarez
- 0000 0004 0512 8863grid.417423.7Laureate Institute for Brain Research, Tulsa, OK USA
| |
Collapse
|
14
|
Abstract
Characterized by the switch of manic and depressive phases, bipolar disorder was described as early as the fifth century BC. Nevertheless up to date, the underlying neurobiology is still largely unclear, assuming a multifactor genesis with both biological-genetic and psychosocial factors. Significant process has been achieved in recent years in researching the causes of bipolar disorder with modern molecular biological (e.g., genetic and epigenetic studies) and imaging techniques (e.g., positron emission tomography (PET) and functional magnetic resonance imaging (fMRI)). In this chapter we will first summarize our recent knowledge on the etiology of bipolar disorder. We then discuss how several factors observed to contribute to bipolar disorder in human patients can be manipulated to generate rodent models for bipolar disorder. Finally, we will give an overview on behavioral test that can be used to assess bipolar-disorder-like behavior in rodents.
Collapse
Affiliation(s)
- Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University, Bochum, Germany.
| | - Georg Juckel
- Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University, Bochum, Germany
| |
Collapse
|
15
|
Bastos JR, Perico KM, Marciano Vieira ÉL, Teixeira AL, Machado FS, de Miranda AS, Moreira FA. Inhibition of the dopamine transporter as an animal model of bipolar disorder mania: Locomotor response, neuroimmunological profile and pharmacological modulation. J Psychiatr Res 2018; 102:142-149. [PMID: 29656188 DOI: 10.1016/j.jpsychires.2018.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/29/2018] [Accepted: 04/05/2018] [Indexed: 12/22/2022]
Abstract
Inhibition of dopamine transporter (DAT) by GBR12909 has been proposed as a pharmacological model of mania related to bipolar disorder (BD). Here we tested the hypothesis that GBR12909 injection impairs habituation and induces hyperlocomotion in mice, along with changes in cytokines and neurotrophic factors levels, as observed in BD patients. We also tested if lithium carbonate, sodium valproate and aripiprazole prevent GBR12909-induced locomotion. Male Swiss mice received GBR12909 (15 mg/kg) injections and locomotor responses were quantified in an open field. Cytokines and neurotrophic factors levels were assessed in the prefrontal cortex, striatum and hippocampus 30 min and 24 h after injections. Pre-treatments with lithium, valproate or aripiprazole were performed with single and repeated injection protocols. GBR12909 prevented motoric habituation and increased basal locomotion in habituated mice in the open field. This compound also induced changes in IL-2 and BDNF levels in prefrontal cortex; IL-2, IL-4 and IL-10 in striatum; and IL-10, IL-4, IFN-γ and NGF in hippocampus. GBR12909-induced hyperlocomotion was attenuated by lithium (12.5-100 mg/kg), but not valproate (75-300 mg/kg), and prevented by aripiprazole (0.1-10 mg/kg). Repeated injections of these drugs (twice a day for 3 days), however, failed to inhibit hyperlocomotion. The main limitations of the protocols in this study are the analysis of locomotion as the only behavioral parameter, changes in immune factors that may overlap with other psychiatric disorders and the lack chronic drug injections. Despite of these limitations, this study adds to previous literature suggesting DAT inhibition as a potential animal model of mania related to BD.
Collapse
Affiliation(s)
- Juliana R Bastos
- Grad School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Katherinne M Perico
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Érica L Marciano Vieira
- Grad School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Universidade Federal de Minas Gerais, Brazil
| | - Antônio L Teixeira
- Grad School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Universidade Federal de Minas Gerais, Brazil; Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fabiana S Machado
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Aline S de Miranda
- Grad School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Universidade Federal de Minas Gerais, Brazil; Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Fabrício A Moreira
- Grad School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
16
|
Ashbrook DG, Mulligan MK, Williams RW. Post-genomic behavioral genetics: From revolution to routine. GENES, BRAIN, AND BEHAVIOR 2018; 17:e12441. [PMID: 29193773 PMCID: PMC5876106 DOI: 10.1111/gbb.12441] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/02/2017] [Accepted: 11/20/2017] [Indexed: 12/16/2022]
Abstract
What was once expensive and revolutionary-full-genome sequence-is now affordable and routine. Costs will continue to drop, opening up new frontiers in behavioral genetics. This shift in costs from the genome to the phenome is most notable in large clinical studies of behavior and associated diseases in cohorts that exceed hundreds of thousands of subjects. Examples include the Women's Health Initiative (www.whi.org), the Million Veterans Program (www. RESEARCH va.gov/MVP), the 100 000 Genomes Project (genomicsengland.co.uk) and commercial efforts such as those by deCode (www.decode.com) and 23andme (www.23andme.com). The same transition is happening in experimental neuro- and behavioral genetics, and sample sizes of many hundreds of cases are becoming routine (www.genenetwork.org, www.mousephenotyping.org). There are two major consequences of this new affordability of massive omics datasets: (1) it is now far more practical to explore genetic modulation of behavioral differences and the key role of gene-by-environment interactions. Researchers are already doing the hard part-the quantitative analysis of behavior. Adding the omics component can provide powerful links to molecules, cells, circuits and even better treatment. (2) There is an acute need to highlight and train behavioral scientists in how best to exploit new omics approaches. This review addresses this second issue and highlights several new trends and opportunities that will be of interest to experts in animal and human behaviors.
Collapse
Affiliation(s)
- D G Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Sciences Center, College of Medicine, Memphis, Tennessee
| | - M K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Sciences Center, College of Medicine, Memphis, Tennessee
| | - R W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Sciences Center, College of Medicine, Memphis, Tennessee
| |
Collapse
|
17
|
Minassian A, Young JW, Geyer MA, Kelsoe JR, Perry W. The COMT Val158Met Polymorphism and Exploratory Behavior in Bipolar Mania. MOLECULAR NEUROPSYCHIATRY 2018; 3:151-156. [PMID: 29594134 PMCID: PMC5836247 DOI: 10.1159/000481822] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/12/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The catechol-O-methyltransferase (COMT) Val158Met gene influences cognition and behavior in psychiatric illnesses; its low-activity allele, methionine (Met), may be associated with behavior reflecting catecholamine overactivity. Heightened motor activity and increased positive valence are central features of bipolar disorder (BD) and have been quantified in the human Behavioral Pattern Monitor (hBPM), an exploration paradigm based upon the rodent open field. We examined whether hBPM behavior was related to the COMT gene in a small sample of manic BD patients. METHODS Twenty-six acutely hospitalized manic BD patients were genotyped for the COMT Val158Met polymorphism and tested in the hBPM, an unfamiliar room containing novel objects. Movements around the hBPM and object interactions were video-recorded for 15 min and rated. RESULTS Met homozygote BD patients demonstrated significantly more interactions with multiple objects and more time spent with objects in the hBPM. Valine (Val) homozygote patients exhibited the least object exploration, while heterozygote patients demonstrated intermediate levels. CONCLUSION This preliminary study suggests that arousal and positive valence are influenced in a linear fashion by COMT, presumably due to increased catecholamine in frontal regions, but these findings require replication in a larger sample. The hBPM can enable cross-species and transdiagnostic studies to inform neurobiology of psychiatric disorders.
Collapse
Affiliation(s)
- Arpi Minassian
- *Arpi Minassian, PhD, Department of Psychiatry, University of California San Diego, 200 West Arbor Drive, Mailcode 8620, San Diego, CA 92109–8620 (USA), E-Mail
| | | | | | | | | |
Collapse
|
18
|
Abstract
Because of the ethical and regulatory hurdles associated with human studies, much of what is known about the psychopharmacology of hallucinogens has been derived from animal models. However, developing reliable animal models has proven to be a challenging task due to the complexity and variability of hallucinogen effects in humans. This chapter focuses on three animal models that are frequently used to test the effects of hallucinogens on unconditioned behavior: head twitch response (HTR), prepulse inhibition of startle (PPI), and exploratory behavior. The HTR has demonstrated considerable utility in the neurochemical actions of hallucinogens. However, the latter two models have clearer conceptual bridges to human phenomenology. Consistent with the known mechanism of action of hallucinogens in humans, the behavioral effects of hallucinogens in rodents are mediated primarily by activation of 5-HT2A receptors. There is evidence, however, that other receptors may play secondary roles. The structure-activity relationships (SAR) of hallucinogens are reviewed in relation to each model, with a focus on the HTR in rats and mice.
Collapse
Affiliation(s)
- Adam L Halberstadt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093-0804, USA.
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093-0804, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
19
|
Pappas AL, Bey AL, Wang X, Rossi M, Kim YH, Yan H, Porkka F, Duffney LJ, Phillips SM, Cao X, Ding JD, Rodriguiz RM, Yin HH, Weinberg RJ, Ji RR, Wetsel WC, Jiang YH. Deficiency of Shank2 causes mania-like behavior that responds to mood stabilizers. JCI Insight 2017; 2:92052. [PMID: 29046483 PMCID: PMC5846902 DOI: 10.1172/jci.insight.92052] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/06/2017] [Indexed: 12/22/2022] Open
Abstract
Genetic defects in the synaptic scaffolding protein gene, SHANK2, are linked to a variety of neuropsychiatric disorders, including autism spectrum disorders, schizophrenia, intellectual disability, and bipolar disorder, but the molecular mechanisms underlying the pleotropic effects of SHANK2 mutations are poorly understood. We generated and characterized a line of Shank2 mutant mice by deleting exon 24 (Δe24). Shank2Δe24-/- mice engage in significantly increased locomotor activity, display abnormal reward-seeking behavior, are anhedonic, have perturbations in circadian rhythms, and show deficits in social and cognitive behaviors. While these phenotypes recapitulate the pleotropic behaviors associated with human SHANK2-related disorders, major behavioral features in these mice are reminiscent of bipolar disorder. For instance, their hyperactivity was augmented with amphetamine but was normalized with the mood stabilizers lithium and valproate. Shank2 deficiency limited to the forebrain recapitulated the bipolar mania phenotype. The composition and functions of NMDA and AMPA receptors were altered at Shank2-deficient synapses, hinting toward the mechanism underlying these behavioral abnormalities. Human genetic findings support construct validity, and the behavioral features in Shank2 Δe24 mice support face and predictive validities of this model for bipolar mania. Further genetic studies to understand the contribution of SHANK2 deficiencies in bipolar disorder are warranted.
Collapse
Affiliation(s)
- Andrea L. Pappas
- Department of Neurobiology
- Cellular and Molecular Biology Program
| | | | | | | | | | | | - Fiona Porkka
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
| | | | | | | | - Jin-dong Ding
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ramona M. Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
| | - Henry H. Yin
- Department of Neurobiology
- Department of Psychology and Neuroscience
| | - Richard J. Weinberg
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ru-Rong Ji
- Department of Neurobiology
- Cellular and Molecular Biology Program
- Department of Anesthesiology, and
| | - William C. Wetsel
- Department of Neurobiology
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
- Department of Cell Biology
- Duke Institute of Brain Science, and
| | - Yong-hui Jiang
- Department of Neurobiology
- Cellular and Molecular Biology Program
- Department of Pediatrics
- Duke Institute of Brain Science, and
- Genomics and Genetics Graduate Program, Duke University, Durham, North Carolina, USA
| |
Collapse
|
20
|
Beyer DKE, Freund N. Animal models for bipolar disorder: from bedside to the cage. Int J Bipolar Disord 2017; 5:35. [PMID: 29027157 PMCID: PMC5638767 DOI: 10.1186/s40345-017-0104-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/11/2017] [Indexed: 12/28/2022] Open
Abstract
Bipolar disorder is characterized by recurrent manic and depressive episodes. Patients suffering from this disorder experience dramatic mood swings with a wide variety of typical behavioral facets, affecting overall activity, energy, sexual behavior, sense of self, self-esteem, circadian rhythm, cognition, and increased risk for suicide. Effective treatment options are limited and diagnosis can be complicated. To overcome these obstacles, a better understanding of the neurobiology underlying bipolar disorder is needed. Animal models can be useful tools in understanding brain mechanisms associated with certain behavior. The following review discusses several pathological aspects of humans suffering from bipolar disorder and compares these findings with insights obtained from several animal models mimicking diverse facets of its symptomatology. Various sections of the review concentrate on specific topics that are relevant in human patients, namely circadian rhythms, neurotransmitters, focusing on the dopaminergic system, stressful environment, and the immune system. We then explain how these areas have been manipulated to create animal models for the disorder. Even though several approaches have been conducted, there is still a lack of adequate animal models for bipolar disorder. Specifically, most animal models mimic only mania or depression and only a few include the cyclical nature of the human condition. Future studies could therefore focus on modeling both episodes in the same animal model to also have the possibility to investigate the switch from mania-like behavior to depressive-like behavior and vice versa. The use of viral tools and a focus on circadian rhythms and the immune system might make the creation of such animal models possible.
Collapse
Affiliation(s)
- Dominik K. E. Beyer
- Experimental and Molecular Psychiatry, LWL University Hospital, Ruhr University Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| | - Nadja Freund
- Experimental and Molecular Psychiatry, LWL University Hospital, Ruhr University Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| |
Collapse
|
21
|
Milienne-Petiot M, Groenink L, Minassian A, Young JW. Blockade of dopamine D 1-family receptors attenuates the mania-like hyperactive, risk-preferring, and high motivation behavioral profile of mice with low dopamine transporter levels. J Psychopharmacol 2017; 31:1334-1346. [PMID: 28950781 PMCID: PMC10773978 DOI: 10.1177/0269881117731162] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with bipolar disorder mania exhibit poor cognition, impulsivity, risk-taking, and goal-directed activity that negatively impact their quality of life. To date, existing treatments for bipolar disorder do not adequately remediate cognitive dysfunction. Reducing dopamine transporter expression recreates many bipolar disorder mania-relevant behaviors (i.e. hyperactivity and risk-taking). The current study investigated whether dopamine D1-family receptor blockade would attenuate the risk-taking, hypermotivation, and hyperactivity of dopamine transporter knockdown mice. METHODS Dopamine transporter knockdown and wild-type littermate mice were tested in mouse versions of the Iowa Gambling Task (risk-taking), Progressive Ratio Breakpoint Test (effortful motivation), and Behavioral Pattern Monitor (activity). Prior to testing, the mice were treated with the dopamine D1-family receptor antagonist SCH 23390 hydrochloride (0.03, 0.1, or 0.3 mg/kg), or vehicle. RESULTS Dopamine transporter knockdown mice exhibited hyperactivity and hyperexploration, hypermotivation, and risk-taking preference compared with wild-type littermates. SCH 23390 hydrochloride treatment decreased premature responding in dopamine transporter knockdown mice and attenuated their hypermotivation. SCH 23390 hydrochloride flattened the safe/risk preference, while reducing activity and exploratory levels of both genotypes similarly. CONCLUSIONS Dopamine transporter knockdown mice exhibited mania-relevant behavior compared to wild-type mice. Systemic dopamine D1-family receptor antagonism attenuated these behaviors in dopamine transporter knockdown, but not all effects were specific to only the knockdown mice. The normalization of behavior via blockade of dopamine D1-family receptors supports the hypothesis that D1 and/or D5 receptors could contribute to the mania-relevant behaviors of dopamine transporter knockdown mice.
Collapse
Affiliation(s)
- Morgane Milienne-Petiot
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States of America
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Lucianne Groenink
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Arpi Minassian
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States of America
| | - Jared W. Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States of America
- Research Service, VA San Diego Healthcare System, San Diego, CA, United States of America
| |
Collapse
|
22
|
Kirlic N, Aupperle RL, Misaki M, Kuplicki R, Alvarez RP. Recruitment of orbitofrontal cortex during unpredictable threat among adults at risk for affective disorders. Brain Behav 2017; 7:e00757. [PMID: 28828218 PMCID: PMC5561318 DOI: 10.1002/brb3.757] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 05/01/2017] [Accepted: 06/03/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Mood and anxiety disorders are characterized by altered prefrontal-amygdala function and increased behavioral inhibition (BI) in response to potential threat. Whether these alterations constitute a vulnerability or a symptom of illness remains unclear. The medial orbitofrontal cortex (mOFC) is thought to play a central role in estimating probability and cost of threat, in turn informing selection of subsequent behaviors. To better understand the behavioral and neural processes that may be associated with risk for psychopathology, we used a virtual reality paradigm to examine behavioral and neural responses of psychiatrically healthy adults with familial history of affective disorders during anticipation of unpredictable threat. METHODS Twenty psychiatrically healthy adults with high familial risk for affective disorders and 20 low-risk matched controls underwent functional magnetic resonance imaging concurrent with a paradigm in which they explored virtual contexts associated with the threat of shock or safety from shock. Subjective anxiety ratings, skin conductance, exploratory behavior, and neural responses were examined for threat versus safe conditions. RESULTS High-risk adults evidenced greater right mOFC activation, as well as greater BI, compared to low-risk adults. There were no significant group differences in subjective ratings or autonomic responses. Individuals exhibiting greater activity in the right mOFC showed greater BI and decreased skin conductance response. CONCLUSIONS These results suggest that BI and mOFC recruitment during anticipation of aversive outcomes may reflect a vulnerability for affective disorders. However, such a response may also serve as a compensatory response, protecting these high-risk individuals from negative outcomes (i.e., increased physiological arousal). These results suggest that the OFC may play a central role in driving threat-related behaviors and thus may be a target for efforts aimed at early detection or prevention.
Collapse
Affiliation(s)
- Namik Kirlic
- Laureate Institute for Brain Research Tulsa OK USA
| | - Robin L Aupperle
- Laureate Institute for Brain Research Tulsa OK USA.,Department of Community Medicine University of Tulsa Tulsa OK USA
| | | | | | | |
Collapse
|
23
|
Cope ZA, Minassian A, Kreitner D, MacQueen DA, Milienne-Petiot M, Geyer MA, Perry W, Young JW. Modafinil improves attentional performance in healthy, non-sleep deprived humans at doses not inducing hyperarousal across species. Neuropharmacology 2017; 125:254-262. [PMID: 28774856 DOI: 10.1016/j.neuropharm.2017.07.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/27/2017] [Accepted: 07/30/2017] [Indexed: 12/18/2022]
Abstract
The wake-promoting drug modafinil is frequently used off-label to improve cognition in psychiatric and academic populations alike. The domain-specific attentional benefits of modafinil have yet to be quantified objectively in healthy human volunteers using tasks validated for comparison across species. Further, given that modafinil is a low-affinity inhibitor for the dopamine and norepinephrine transporters (DAT/NET respectively) it is unclear if any effects are attributable to a non-specific increase in arousal, a feature of many catecholamine reuptake inhibitors (e.g., cocaine, amphetamine). These experiments were designed to test for domain-specific enhancement of attention and cognitive control by modafinil (200 and 400 mg) in healthy volunteers using the 5-choice continuous performance task (5C-CPT) and Wisconsin Card Sort Task (WCST). An additional cross-species assessment of arousal and hyperactivity was performed in this group and in mice (3.2, 10, or 32 mg/kg) using species-specific versions of the behavioral pattern monitor (BPM). Modafinil significantly enhanced attention (d prime) in humans performing the 5C-CPT at doses that did not affect WCST performance or induce hyperactivity in the BPM. In mice, only the highest dose elicited increased activity in the BPM. These results indicate that modafinil produces domain-specific enhancement of attention in humans not driven by hyperarousal, unlike other drugs in this class, and higher equivalent doses were required for hyperarousal in mice. Further, these data support the utility of using the 5C-CPT across species to more precisely determine the mechanism(s) underlying the pro-cognitive effects of modafinil and potentially other pharmacological treatments.
Collapse
Affiliation(s)
- Zackary A Cope
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States
| | - Arpi Minassian
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Center for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA, United States
| | - Dustin Kreitner
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States
| | - David A MacQueen
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States
| | - Morgane Milienne-Petiot
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, David de Wied Building, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Mark A Geyer
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States
| | - William Perry
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States
| | - Jared W Young
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States.
| |
Collapse
|
24
|
Kirlic N, Young J, Aupperle RL. Animal to human translational paradigms relevant for approach avoidance conflict decision making. Behav Res Ther 2017; 96:14-29. [PMID: 28495358 DOI: 10.1016/j.brat.2017.04.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 04/17/2017] [Accepted: 04/20/2017] [Indexed: 12/29/2022]
Abstract
Avoidance behavior in clinical anxiety disorders is often a decision made in response to approach-avoidance conflict, resulting in a sacrifice of potential rewards to avoid potential negative affective consequences. Animal research has a long history of relying on paradigms related to approach-avoidance conflict to model anxiety-relevant behavior. This approach includes punishment-based conflict, exploratory, and social interaction tasks. There has been a recent surge of interest in the translation of paradigms from animal to human, in efforts to increase generalization of findings and support the development of more effective mental health treatments. This article briefly reviews animal tests related to approach-avoidance conflict and results from lesion and pharmacologic studies utilizing these tests. We then provide a description of translational human paradigms that have been developed to tap into related constructs, summarizing behavioral and neuroimaging findings. Similarities and differences in findings from analogous animal and human paradigms are discussed. Lastly, we highlight opportunities for future research and paradigm development that will support the clinical utility of this translational work.
Collapse
Affiliation(s)
- Namik Kirlic
- Laureate Institute for Brain Research, 6655 S Yale Ave, Tulsa, OK 74136, United States.
| | - Jared Young
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093, United States; VA San Diego Healthcare System, 3350 La Jolla Village Dr, San Diego, CA 92161, United States.
| | - Robin L Aupperle
- Laureate Institute for Brain Research, 6655 S Yale Ave, Tulsa, OK 74136, United States; School of Community Medicine, University of Tulsa, 800 S Tucker Dr, Tulsa, OK 74104, United States.
| |
Collapse
|
25
|
Milienne-Petiot M, Geyer MA, Arnt J, Young JW. Brexpiprazole reduces hyperactivity, impulsivity, and risk-preference behavior in mice with dopamine transporter knockdown-a model of mania. Psychopharmacology (Berl) 2017; 234:1017-1028. [PMID: 28160035 PMCID: PMC5391249 DOI: 10.1007/s00213-017-4543-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
RATIONALE Bipolar disorder (BD) is a unique mood disorder defined by periods of depression and mania. The defining diagnosis of BD is the presence of mania/hypomania, with symptoms including hyperactivity and risk-taking. Since current treatments do not ameliorate cognitive deficits such as risky decision-making, and impulsivity that can negatively affect a patient's quality of life, better treatments are needed. OBJECTIVES Here, we tested whether acute treatment with brexpiprazole, a serotonin-dopamine activity modulator with partial agonist activity at D2/3 and 5-HT1A receptors, would attenuate the BD mania-relevant behaviors of the dopamine transporter (DAT) knockdown mouse model of mania. METHODS The effects of brexpiprazole on DAT knockdown and wild-type littermate mice were examined in the behavioral pattern monitor (BPM) and Iowa gambling task (IGT) to quantify activity/exploration and impulsivity/risk-taking behavior respectively. RESULTS DAT knockdown mice exhibited hyper-exploratory behavior in the BPM and made fewer safe choices in the IGT. Brexpiprazole attenuated the mania-like hyper-exploratory phenotype and increased safe choices in risk-preferring DAT knockdown mice. Brexpiprazole also reduced safe choices in safe-preferring mice irrespective of genotype. Finally, brexpiprazole reduced premature (impulsive-like) responses in both groups of mice. CONCLUSIONS Consistent with earlier reports, DAT knockdown mice exhibited hyper-exploratory, risk-preferring, and impulsive-like profiles consistent with patients with BD mania in these tasks. These behaviors were attenuated after brexpiprazole treatment. These data therefore indicate that brexpiprazole could be a novel treatment for BD mania and/or risk-taking/impulsivity disorders, since it remediates some relevant behavioral abnormalities in this mouse model.
Collapse
Affiliation(s)
- Morgane Milienne-Petiot
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, The Netherlands
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Jørn Arnt
- Sunred Pharma Consulting, Solrod Strand, Denmark
- Synaptic Transmission, Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, DK, Denmark
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, USA.
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
26
|
Palmius N, Tsanas A, Saunders KEA, Bilderbeck AC, Geddes JR, Goodwin GM, De Vos M. Detecting Bipolar Depression From Geographic Location Data. IEEE Trans Biomed Eng 2016; 64:1761-1771. [PMID: 28113247 PMCID: PMC5947818 DOI: 10.1109/tbme.2016.2611862] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective This paper aims to identify periods of depression using geolocation movements recorded from mobile phones in a prospective community study of individuals with bipolar disorder (BD). Methods Anonymized geographic location recordings from 22 BD participants and 14 healthy controls (HC) were collected over 3 months. Participants reported their depressive symptomatology using a weekly questionnaire (QIDS-SR16). Recorded location data were preprocessed by detecting and removing imprecise data points and features were extracted to assess the level and regularity of geographic movements of the participant. A subset of features were selected using a wrapper feature selection method and presented to 1) a linear regression model and a quadratic generalized linear model with a logistic link function for questionnaire score estimation; and 2) a quadratic discriminant analysis classifier for depression detection in BD participants based on their questionnaire responses. Results HC participants did not report depressive symptoms and their features showed similar distributions to nondepressed BD participants. Questionnaire score estimation using geolocation-derived features from BD participants demonstrated an optimal mean absolute error rate of 3.73, while depression detection demonstrated an optimal (median ± IQR) F1 score of 0.857 ± 0.022 using five features (classification accuracy: 0.849 ± 0.016; sensitivity: 0.839 ± 0.014; specificity: 0.872 ± 0.047). Conclusion These results demonstrate a strong link between geographic movements and depression in bipolar disorder. Significance To our knowledge, this is the first community study of passively recorded objective markers of depression in bipolar disorder of this scale. The techniques could help individuals monitor their depression and enable healthcare providers to detect those in need of care or treatment.
Collapse
|
27
|
Schoenrock SA, Tarantino LM. Developmental vitamin D deficiency and schizophrenia: the role of animal models. GENES BRAIN AND BEHAVIOR 2016; 15:45-61. [PMID: 26560996 DOI: 10.1111/gbb.12271] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/22/2015] [Accepted: 11/04/2015] [Indexed: 01/10/2023]
Abstract
Schizophrenia is a debilitating neuropsychiatric disorder that affects 1% of the US population. Based on twin and genome-wide association studies, it is clear that both genetics and environmental factors increase the risk for developing schizophrenia. Moreover, there is evidence that conditions in utero, either alone or in concert with genetic factors, may alter neurodevelopment and lead to an increased risk for schizophrenia. There has been progress in identifying genetic loci and environmental exposures that increase risk, but there are still considerable gaps in our knowledge. Furthermore, very little is known about the specific neurodevelopmental mechanisms upon which genetics and the environment act to increase disposition to developing schizophrenia in adulthood. Vitamin D deficiency during the perinatal period has been hypothesized to increase risk for schizophrenia in humans. The developmental vitamin D (DVD) deficiency hypothesis of schizophrenia arises from the observation that disease risk is increased in individuals who are born in winter or spring, live further from the equator or live in urban vs. rural settings. These environments result in less exposure to sunlight, thereby reducing the initial steps in the production of vitamin D. Rodent models have been developed to characterize the behavioral and developmental effects of DVD deficiency. This review focuses on these animal models and discusses the current knowledge of the role of DVD deficiency in altering behavior and neurobiology relevant to schizophrenia.
Collapse
Affiliation(s)
- S A Schoenrock
- Department of Psychiatry, School of Medicine, Chapel Hill, NC, USA.,Neurobiology Curriculum, Chapel Hill, NC, USA
| | - L M Tarantino
- Department of Psychiatry, School of Medicine, Chapel Hill, NC, USA.,Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
28
|
Soontornniyomkij V, Kesby JP, Morgan EE, Bischoff-Grethe A, Minassian A, Brown GG, Grant I. Effects of HIV and Methamphetamine on Brain and Behavior: Evidence from Human Studies and Animal Models. J Neuroimmune Pharmacol 2016; 11:495-510. [PMID: 27484318 PMCID: PMC4985024 DOI: 10.1007/s11481-016-9699-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/26/2016] [Indexed: 10/21/2022]
Abstract
Methamphetamine (Meth) use is frequent among HIV-infected persons. Combined HIV and Meth insults may exacerbate neural injury in vulnerable neuroanatomic structures or circuitries in the brain, leading to increased behavioral disturbance and cognitive impairment. While acute and chronic effects of Meth in humans and animal models have been studied for decades, the neurobehavioral effects of Meth in the context of HIV infection are much less explored. In-depth understanding of the scope of neurobehavioral phenotypes and mechanisms in HIV/Meth intersection is needed. The present report summarizes published research findings, as well as unpublished data, in humans and animal models with regard to neurobehavioral disturbance, neuroimaging, and neuropathology, and in vitro experimental systems, with an emphasis on findings emerging from the National Institute on Drug Abuse (NIDA) funded Translational Methamphetamine AIDS Research Center (TMARC). Results from human studies and animal (primarily HIV-1 gp120 transgenic mouse) models thus far suggest that combined HIV and Meth insults increase the likelihood of neural injury in the brain. The neurobehavioral effects include cognitive impairment and increased tendencies toward impaired behavioral inhibition and social cognition. These impairments are relevant to behaviors that affect personal and social risks, e.g. worse medication adherence, riskier behaviors, and greater likelihood of HIV transmission. The underlying mechanisms may include electrochemical changes in neuronal circuitries, injury to white matter microstructures, synaptodendritic damage, and selective neuronal loss. Utilization of research methodologies that are valid across species is instrumental in generating new knowledge with clinical translational value.
Collapse
Affiliation(s)
- Virawudh Soontornniyomkij
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA.
| | - James P Kesby
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
- Queensland Brain Institute, The University of Queensland, St. Lucia, Qld, Australia
| | - Erin E Morgan
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| | - Amanda Bischoff-Grethe
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| | - Arpi Minassian
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| | - Gregory G Brown
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| | - Igor Grant
- Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0603, USA
| |
Collapse
|
29
|
Souza LS, Silva EF, Santos WB, Asth L, Lobão-Soares B, Soares-Rachetti VP, Medeiros IU, Gavioli EC. Lithium and valproate prevent methylphenidate-induced mania-like behaviors in the hole board test. Neurosci Lett 2016; 629:143-148. [PMID: 27353513 DOI: 10.1016/j.neulet.2016.06.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022]
Abstract
Manic bipolar is diagnosed by psychomotor agitation, increased goal-directed activity, insomnia, grandiosity, excessive speech, and risky behavior. Animal studies aimed to modeling mania are commonly based in psychostimulants-induced hyperlocomotion. The exploration of other behaviors related with mania is mandatory to investigate this phase of bipolar disorder in animals. In this study, the hole board apparatus was suggested for evaluating mania-like behaviors induced by the psychostimulant methylphenidate. The treatment with methylphenidate (10mg/kg, ip) increased locomotion in the open field test. The pretreatment with lithium (50mg/kg, ip) and valproate (400mg/kg, ip) significantly prevented the hyperlocomotion. In the hole-board test, methylphenidate increased interactions with the central and peripheral holes and the exploration of central areas. Lithium was more effective than valproate in preventing all the behavioral manifestations induced by the psychostimulant. These findings were discussed based on the ability of methylphenidate-treated mice mimicking two symptoms of mania in the hole board test: goal-directed action and risk-taking behavior. In conclusion, the results point to a new approach to study mania through the hole board apparatus. The hole board test appears to be a sensitive assay to detect the efficacy of antimanic drugs.
Collapse
Affiliation(s)
- L S Souza
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - E F Silva
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - W B Santos
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - L Asth
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - B Lobão-Soares
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - V P Soares-Rachetti
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - I U Medeiros
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - E C Gavioli
- Behavioral Pharmacology Laboratory, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
30
|
Zhang Z, Zheng F, You Y, Ma Y, Lu T, Yue W, Zhang D. Growth arrest specific gene 7 is associated with schizophrenia and regulates neuronal migration and morphogenesis. Mol Brain 2016; 9:54. [PMID: 27189492 PMCID: PMC4870797 DOI: 10.1186/s13041-016-0238-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 05/10/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Schizophrenia is a highly heritable chronic mental disorder with significant abnormalities in brain function. The neurodevelopmental hypothesis proposes that schizophrenia originates in the prenatal period due to impairments in neuronal developmental processes such as migration and arborization, leading to abnormal brain maturation. Previous studies have identified multiple promising candidate genes that drive functions in neurodevelopment and are associated with schizophrenia. However, the molecular mechanisms of how they exert effects on the pathophysiology of schizophrenia remain largely unknown. RESULTS In our research, we identified growth arrest specific gene 7 (GAS7) as a schizophrenia risk gene in two independent Han Chinese populations using a two-stage association study. Functional experiments were done to further explore the underlying mechanisms of the role of Gas7 in cortical development. In vitro, we discovered that Gas7 contributed to neurite outgrowth through the F-BAR domain. In vivo, overexpression of Gas7 arrested neuronal migration by increasing leading process branching, while suppression of Gas7 could inhibit neuronal migration by lengthening leading processes. Through a series of behavioral tests, we also found that Gas7-deficient mice showed sensorimotor gating deficits. CONCLUSIONS Our results demonstrate GAS7 as a susceptibility gene for schizophrenia. Gas7 might participate in the pathogenesis of schizophrenia by regulating neurite outgrowth and neuronal migration through its C-terminal F-BAR domain. The impaired pre-pulse inhibition (PPI) of Gas7-deficient mice might mirror the disease-related behavior in schizophrenia.
Collapse
Affiliation(s)
- Zhengrong Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, 51 Hua Yuan Bei Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, 100191, China
| | - Fanfan Zheng
- Institute of Mental Health, The Sixth Hospital, Peking University, 51 Hua Yuan Bei Road, Hai Dian District, Beijing, 100191, China. .,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, 100191, China. .,Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, 95 Zhong Guan Cun East Road, Hai Dian District, Beijing, 100190, China.
| | - Yang You
- Institute of Mental Health, The Sixth Hospital, Peking University, 51 Hua Yuan Bei Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, 100191, China
| | - Yuanlin Ma
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Tianlan Lu
- Institute of Mental Health, The Sixth Hospital, Peking University, 51 Hua Yuan Bei Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, 100191, China
| | - Weihua Yue
- Institute of Mental Health, The Sixth Hospital, Peking University, 51 Hua Yuan Bei Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, 100191, China
| | - Dai Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, 51 Hua Yuan Bei Road, Hai Dian District, Beijing, 100191, China. .,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders (Peking University), Beijing, 100191, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China. .,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
31
|
Sharma AN, Fries GR, Galvez JF, Valvassori SS, Soares JC, Carvalho AF, Quevedo J. Modeling mania in preclinical settings: A comprehensive review. Prog Neuropsychopharmacol Biol Psychiatry 2016; 66:22-34. [PMID: 26545487 PMCID: PMC4728043 DOI: 10.1016/j.pnpbp.2015.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/29/2015] [Accepted: 11/03/2015] [Indexed: 12/17/2022]
Abstract
The current pathophysiological understanding of mechanisms leading to onset and progression of bipolar manic episodes remains limited. At the same time, available animal models for mania have limited face, construct, and predictive validities. Additionally, these models fail to encompass recent pathophysiological frameworks of bipolar disorder (BD), e.g. neuroprogression. Therefore, there is a need to search for novel preclinical models for mania that could comprehensively address these limitations. Herein we review the history, validity, and caveats of currently available animal models for mania. We also review new genetic models for mania, namely knockout mice for genes involved in neurotransmission, synapse formation, and intracellular signaling pathways. Furthermore, we review recent trends in preclinical models for mania that may aid in the comprehension of mechanisms underlying the neuroprogressive and recurring nature of BD. In conclusion, the validity of animal models for mania remains limited. Nevertheless, novel (e.g. genetic) animal models as well as adaptation of existing paradigms hold promise.
Collapse
Affiliation(s)
- Ajaykumar N. Sharma
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Gabriel R. Fries
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Juan F. Galvez
- Department of Psychiatry, Pontificia Universidad Javeriana School of Medicine, Bogotá, Colombia
| | - Samira S. Valvassori
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Jair C. Soares
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - André F. Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Joao Quevedo
- Center for Translational Psychiatry, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil.
| |
Collapse
|
32
|
Kupper Z, Ramseyer F, Hoffmann H, Tschacher W. Nonverbal Synchrony in Social Interactions of Patients with Schizophrenia Indicates Socio-Communicative Deficits. PLoS One 2015; 10:e0145882. [PMID: 26716444 PMCID: PMC4696745 DOI: 10.1371/journal.pone.0145882] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 12/09/2015] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Disordered interpersonal communication can be a serious problem in schizophrenia. Recent advances in computer-based measures allow reliable and objective quantification of nonverbal behavior. Research using these novel measures has shown that objective amounts of body and head movement in patients with schizophrenia during social interactions are closely related to the symptom profiles of these patients. In addition to and above mere amounts of movement, the degree of synchrony, or imitation, between patients and normal interactants may be indicative of core deficits underlying various problems in domains related to interpersonal communication, such as symptoms, social competence, and social functioning. METHODS Nonverbal synchrony was assessed objectively using Motion Energy Analysis (MEA) in 378 brief, videotaped role-play scenes involving 27 stabilized outpatients diagnosed with paranoid-type schizophrenia. RESULTS Low nonverbal synchrony was indicative of symptoms, low social competence, impaired social functioning, and low self-evaluation of competence. These relationships remained largely significant when correcting for the amounts of patients' movement. When patients showed reduced imitation of their interactants' movements, negative symptoms were likely to be prominent. Conversely, positive symptoms were more prominent in patients when their interaction partners' imitation of their movements was reduced. CONCLUSIONS Nonverbal synchrony can be an objective and sensitive indicator of the severity of patients' problems. Furthermore, quantitative analysis of nonverbal synchrony may provide novel insights into specific relationships between symptoms, cognition, and core communicative problems in schizophrenia.
Collapse
Affiliation(s)
- Zeno Kupper
- Division of Molecular Psychiatry, Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| | - Fabian Ramseyer
- Department for Clinical Psychology and Psychotherapy, University of Bern, Bern, Switzerland
- ARTORG—Gerontechnology and Rehabilitation, University Hospital of Old Age Psychiatry, University of Bern, Bern, Switzerland
| | - Holger Hoffmann
- Center for Psychiatric Rehabilitation, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| | - Wolfgang Tschacher
- Division of Systems Neuroscience of Psychopathology, Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| |
Collapse
|
33
|
Logan RW, McClung CA. Animal models of bipolar mania: The past, present and future. Neuroscience 2015; 321:163-188. [PMID: 26314632 DOI: 10.1016/j.neuroscience.2015.08.041] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 12/19/2022]
Abstract
Bipolar disorder (BD) is the sixth leading cause of disability in the world according to the World Health Organization and affects nearly six million (∼2.5% of the population) adults in the United State alone each year. BD is primarily characterized by mood cycling of depressive (e.g., helplessness, reduced energy and activity, and anhedonia) and manic (e.g., increased energy and hyperactivity, reduced need for sleep, impulsivity, reduced anxiety and depression), episodes. The following review describes several animal models of bipolar mania with a focus on more recent findings using genetically modified mice, including several with the potential of investigating the mechanisms underlying 'mood' cycling (or behavioral switching in rodents). We discuss whether each of these models satisfy criteria of validity (i.e., face, predictive, and construct), while highlighting their strengths and limitations. Animal models are helping to address critical questions related to pathophysiology of bipolar mania, in an effort to more clearly define necessary targets of first-line medications, lithium and valproic acid, and to discover novel mechanisms with the hope of developing more effective therapeutics. Future studies will leverage new technologies and strategies for integrating animal and human data to reveal important insights into the etiology, pathophysiology, and treatment of BD.
Collapse
Affiliation(s)
- R W Logan
- University of Pittsburgh School of Medicine, Department of Psychiatry, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, United States
| | - C A McClung
- University of Pittsburgh School of Medicine, Department of Psychiatry, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, United States.
| |
Collapse
|
34
|
van Enkhuizen J, Geyer MA, Minassian A, Perry W, Henry BL, Young JW. Investigating the underlying mechanisms of aberrant behaviors in bipolar disorder from patients to models: Rodent and human studies. Neurosci Biobehav Rev 2015; 58:4-18. [PMID: 26297513 DOI: 10.1016/j.neubiorev.2015.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/03/2015] [Accepted: 08/16/2015] [Indexed: 11/18/2022]
Abstract
Psychiatric patients with bipolar disorder suffer from states of depression and mania, during which a variety of symptoms are present. Current treatments are limited and neurocognitive deficits in particular often remain untreated. Targeted therapies based on the biological mechanisms of bipolar disorder could fill this gap and benefit patients and their families. Developing targeted therapies would benefit from appropriate animal models which are challenging to establish, but remain a vital tool. In this review, we summarize approaches to create a valid model relevant to bipolar disorder. We focus on studies that use translational tests of multivariate exploratory behavior, sensorimotor gating, decision-making under risk, and attentional functioning to discover profiles that are consistent between patients and rodent models. Using this battery of translational tests, similar behavior profiles in bipolar mania patients and mice with reduced dopamine transporter activity have been identified. Future investigations should combine other animal models that are biologically relevant to the neuropsychiatric disorder with translational behavioral assessment as outlined here. This methodology can be utilized to develop novel targeted therapies that relieve symptoms for more patients without common side effects caused by current treatments.
Collapse
Affiliation(s)
- Jordy van Enkhuizen
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States.
| | - Arpi Minassian
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States
| | - William Perry
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States
| | - Brook L Henry
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
35
|
Ashbrook DG, Williams RW, Lu L, Hager R. A cross-species genetic analysis identifies candidate genes for mouse anxiety and human bipolar disorder. Front Behav Neurosci 2015; 9:171. [PMID: 26190982 PMCID: PMC4486840 DOI: 10.3389/fnbeh.2015.00171] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
Bipolar disorder (BD) is a significant neuropsychiatric disorder with a lifetime prevalence of ~1%. To identify genetic variants underlying BD genome-wide association studies (GWAS) have been carried out. While many variants of small effect associated with BD have been identified few have yet been confirmed, partly because of the low power of GWAS due to multiple comparisons being made. Complementary mapping studies using murine models have identified genetic variants for behavioral traits linked to BD, often with high power, but these identified regions often contain too many genes for clear identification of candidate genes. In the current study we have aligned human BD GWAS results and mouse linkage studies to help define and evaluate candidate genes linked to BD, seeking to use the power of the mouse mapping with the precision of GWAS. We use quantitative trait mapping for open field test and elevated zero maze data in the largest mammalian model system, the BXD recombinant inbred mouse population, to identify genomic regions associated with these BD-like phenotypes. We then investigate these regions in whole genome data from the Psychiatric Genomics Consortium's bipolar disorder GWAS to identify candidate genes associated with BD. Finally we establish the biological relevance and pathways of these genes in a comprehensive systems genetics analysis. We identify four genes associated with both mouse anxiety and human BD. While TNR is a novel candidate for BD, we can confirm previously suggested associations with CMYA5, MCTP1, and RXRG. A cross-species, systems genetics analysis shows that MCTP1, RXRG, and TNR coexpress with genes linked to psychiatric disorders and identify the striatum as a potential site of action. CMYA5, MCTP1, RXRG, and TNR are associated with mouse anxiety and human BD. We hypothesize that MCTP1, RXRG, and TNR influence intercellular signaling in the striatum.
Collapse
Affiliation(s)
- David G Ashbrook
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester Manchester, UK
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, University of Tennessee Memphis, TN, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, University of Tennessee Memphis, TN, USA ; Jiangsu Key Laboratory of Neuroregeneration, Nantong University Nantong, China
| | - Reinmar Hager
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester Manchester, UK
| |
Collapse
|
36
|
Kesby JP, Heaton RK, Young JW, Umlauf A, Woods SP, Letendre SL, Markou A, Grant I, Semenova S. Methamphetamine Exposure Combined with HIV-1 Disease or gp120 Expression: Comparison of Learning and Executive Functions in Humans and Mice. Neuropsychopharmacology 2015; 40:1899-909. [PMID: 25652249 PMCID: PMC4839513 DOI: 10.1038/npp.2015.39] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/15/2015] [Accepted: 01/27/2015] [Indexed: 11/09/2022]
Abstract
Methamphetamine dependence is a common comorbid condition among people living with HIV, and may exacerbate HIV-associated neurocognitive disorders. Animal models of neuroAIDS suggest that the gp120 protein may also cause cognitive impairment. The present work evaluated the separate and combined effects of HIV/gp120 and methamphetamine on learning and executive functions in both humans and transgenic mice. Human participants were grouped by HIV serostatus (HIV+ or HIV-) and lifetime methamphetamine dependence (METH+ or METH-). A neurocognitive test battery included domain-specific assessments of learning and executive functions. Mice (gp120+ and gp120-) were exposed to either a methamphetamine binge (METH+) or saline (METH-), then tested in the attentional-set-shifting task to assess learning and executive functions. In humans, HIV status was associated with significant impairments in learning, but less so for executive functions. The frequency of learning impairments varied between groups, with the greatest impairment observed in the HIV+/METH+ group. In mice, gp120 expression was associated with impairments in learning but not reversal learning (executive component). The greatest proportion of mice that failed to complete the task was observed in the gp120+/METH+ group, suggesting greater learning impairments. Our cross-species study demonstrated that HIV in humans and gp120 in mice impaired learning, and that a history of methamphetamine exposure increased the susceptibility to HIV-associated neurocognitive deficits in both species. Finally, the similar pattern of results in both species suggest that the gp120 protein may contribute to HIV-associated learning deficits in humans.
Collapse
Affiliation(s)
- James P Kesby
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Robert K Heaton
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jared W Young
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA,Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Anya Umlauf
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Steven P Woods
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Scott L Letendre
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Athina Markou
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Igor Grant
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Svetlana Semenova
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA,Department of Psychiatry, School of Medicine, University of California San Diego, 9500 Gilman Drive, M/C 0603, La Jolla, CA 92093, USA, Tel: +858 534 1528, Fax: +858 534 9917, E mail:
| |
Collapse
|
37
|
Pathak G, Ibrahim BA, McCarthy SA, Baker K, Kelly MP. Amphetamine sensitization in mice is sufficient to produce both manic- and depressive-related behaviors as well as changes in the functional connectivity of corticolimbic structures. Neuropharmacology 2015; 95:434-47. [PMID: 25959066 DOI: 10.1016/j.neuropharm.2015.04.026] [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] [Received: 11/15/2014] [Revised: 04/21/2015] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
Abstract
It has been suggested that amphetamine abuse and withdrawal mimics the diverse nature of bipolar disorder symptomatology in humans. Here, we determined if a single paradigm of amphetamine sensitization would be sufficient to produce both manic- and depressive-related behaviors in mice. CD-1 mice were subcutaneously dosed for 5 days with 1.8 mg/kg d-amphetamine or vehicle. On days 6-31 of withdrawal, amphetamine-sensitized (AS) mice were compared to vehicle-treated (VT) mice on a range of behavioral and biochemical endpoints. AS mice demonstrated reliable mania- and depression-related behaviors from day 7 to day 28 of withdrawal. Relative to VT mice, AS mice exhibited long-lasting mania-like hyperactivity following either an acute 30-min restraint stress or a low-dose 1 mg/kg d-amphetamine challenge, which was attenuated by the mood-stabilizers lithium and quetiapine. In absence of any challenge, AS mice showed anhedonia-like decreases in sucrose preference and depression-like impairments in the off-line consolidation of motor memory, as reflected by the lack of spontaneous improvement across days of training on the rotarod. AS mice also demonstrated a functional impairment in nest building, an ethologically-relevant activity of daily living. Western blot analyses revealed a significant increase in methylation of histone 3 at lysine 9 (H3K9), but not lysine 4 (H3K4), in hippocampus of AS mice relative to VT mice. In situ hybridization for the immediate-early gene activity-regulated cytoskeleton-associated protein (Arc) further revealed heightened activation of corticolimbic structures, decreased functional connectivity between frontal cortex and striatum, and increased functional connectivity between the amygdala and hippocampus of AS mice. The effects of amphetamine sensitization were blunted in C57BL/6J mice relative to CD-1 mice. These results show that a single amphetamine sensitization protocol is sufficient to produce behavioral, functional, and biochemical phenotypes in mice that are relevant to bipolar disorder.
Collapse
Affiliation(s)
- G Pathak
- University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - B A Ibrahim
- University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | | | - K Baker
- Pfizer, Neuroscience, Groton, CT 06340, USA
| | - M P Kelly
- University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|
38
|
Investigating the mechanism(s) underlying switching between states in bipolar disorder. Eur J Pharmacol 2015; 759:151-62. [PMID: 25814263 DOI: 10.1016/j.ejphar.2015.03.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/03/2015] [Accepted: 03/12/2015] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a unique disorder that transcends domains of function since the same patient can exhibit depression or mania, states with polar opposite mood symptoms. During depression, people feel helplessness, reduced energy, and risk aversion, while with mania behaviors include grandiosity, increased energy, less sleep, and risk preference. The neural mechanism(s) underlying each state are gaining clarity, with catecholaminergic disruption seen during mania, and cholinergic dysfunction during depression. The fact that the same patient cycles/switches between these states is the defining characteristic of BD however. Of greater importance therefore, is the mechanism(s) underlying cycling from one state - and its associated neural changes - to another, considered the 'holy grail' of BD research. Herein, we review studies investigating triggers that induce switching to these states. By identifying such triggers, researchers can study neural mechanisms underlying each state and importantly how such mechanistic changes can occur in the same subject. Current animal models of this switch are also discussed, from submissive- and dominant-behaviors to kindling effects. Focus however, is placed on how seasonal changes can induce manic and depressive states in BD sufferers. Importantly, changing photoperiod lengths can induce local switches in neurotransmitter expression in normal animals, from increased catecholaminergic expression during periods of high activity, to increased somatostatin and corticotrophin releasing factor during periods of low activity. Identifying susceptibilities to this switch would enable the development of targeted animal models. From animal models, targeted treatments could be developed and tested that would minimize the likelihood of switching.
Collapse
|
39
|
Abrial E, Bétourné A, Etiévant A, Lucas G, Scarna H, Lambás-Señas L, Haddjeri N. Protein kinase C inhibition rescues manic-like behaviors and hippocampal cell proliferation deficits in the sleep deprivation model of mania. Int J Neuropsychopharmacol 2015; 18:pyu031. [PMID: 25577667 PMCID: PMC4368890 DOI: 10.1093/ijnp/pyu031] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Recent studies revealed that bipolar disorder may be associated with deficits of neuroplasticity. Additionally, accumulating evidence has implicated alterations of the intracellular signaling molecule protein kinase C (PKC) in mania. METHODS Using sleep deprivation (SD) as an animal model of mania, this study aimed to examine the possible relationship between PKC and neuroplasticity in mania. Rats were subjected to SD for 72 h and tested behaviorally. In parallel, SD-induced changes in hippocampal cell proliferation were evaluated with bromodeoxyuridine (BrdU) labeling. We then examined the effects of the mood stabilizer lithium, the antipsychotic agent aripiprazole, and the PKC inhibitors chelerythrine and tamoxifen on both behavioral and cell proliferation impairments induced by SD. The antidepressant fluoxetine was used as a negative control. RESULTS We found that SD triggered the manic-like behaviors such as hyperlocomotion and increased sleep latency, and reduced hippocampal cell proliferation. These alterations were counteracted by an acute administration of lithium and aripiprazole but not of fluoxetine, and only a single administration of aripiprazole increased cell proliferation on its own. Importantly, SD rats exhibited increased levels of phosphorylated synaptosomal-associated protein 25 (SNAP-25) in the hippocampus and prefrontal cortex, suggesting PKC overactivity. Moreover, PKC inhibitors attenuated manic-like behaviors and rescued cell proliferation deficits induced by SD. CONCLUSIONS Our findings confirm the relevance of SD as a model of mania, and provide evidence that antimanic agents are also able to prevent SD-induced decrease of hippocampal cell proliferation. Furthermore, they emphasize the therapeutic potential of PKC inhibitors, as revealed by their antimanic-like and pro-proliferative properties.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nasser Haddjeri
- INSERM U846, Stem Cell and Brain Research Institute, F-69500 Bron, France (Drs Abrial, Etiévant, Lucas, Scarna, Lambás-Señas, and Haddjeri); Université de Lyon, Université Lyon 1, F-69373 Lyon, France (Drs Abrial, Etiévant, Lucas, Scarna, Lambás-Señas, and Haddjeri); Centre de Physiopathologie de Toulouse Purpan, INSERM UMR1043/CNRS UMR 5282, Université Toulouse III, CHU Purpan, BP 3028, F-31024 Toulouse, France (Dr Bétourné).
| |
Collapse
|
40
|
Abstract
The quantification of unconditioned motoric activity is one of the oldest and most commonly utilized tools in behavioral studies. Although typically measured in reference to psychiatric disorders, e.g., amphetamine-induced hyperactivity used as a model of schizophrenia, bipolar disorder (BD), and Tourette's syndrome, the motoric behavior of psychiatric patients had not been quantified similarly to rodents until recently. The rodent behavioral pattern monitor (BPM) was reverse-translated for use in humans, providing the quantification of not only motoric activity but also the locomotor exploratory profile of various psychiatric populations. This measurement includes the quantification of specific exploration and locomotor patterns. As an example, patients with BD, schizophrenia, and those with history of methamphetamine dependence exhibited unique locomotor profiles. It was subsequently determined that reducing dopamine transporter function selectively recreated the locomotor profile of BD mania patients and not any other patient population. Hence, multivariate locomotor profiling offers a first-step approach toward understanding the neural mechanism(s) underlying abnormal behavior in patients with psychiatric disorders. Advances in wearable technology will undoubtedly enable similar multivariate assessments of exploratory and locomotor behavior in "real-world" contexts. Furthermore, trans-diagnostic studies of locomotor activity profiles will inform about essential brain-based functions that cut across diagnostic nosologies.
Collapse
|
41
|
Isolation rearing effects on probabilistic learning and cognitive flexibility in rats. COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2014; 14:388-406. [PMID: 23943516 DOI: 10.3758/s13415-013-0204-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Isolation rearing is a neurodevelopmental manipulation that produces neurochemical, structural, and behavioral alterations in rodents that in many ways are consistent with schizophrenia. Symptoms induced by isolation rearing that mirror clinically relevant aspects of schizophrenia, such as cognitive deficits, open up the possibility of testing putative therapeutics in isolation-reared animals prior to clinical development. We investigated what effect isolation rearing would have on cognitive flexibility, a cognitive function characteristically disrupted in schizophrenia. For this purpose, we assessed cognitive flexibility using between- and within-session probabilistic reversal-learning tasks based on clinical tests. Isolation-reared rats required more sessions, though not more task trials, to acquire criterion performance in the reversal phase of the task, and were slower to adjust their task strategy after reward contingencies were switched. Isolation-reared rats also completed fewer trials and exhibited lower levels of overall activity in the probabilistic reversal-learning task than did the socially reared rats. This finding contrasted with the elevated levels of unconditioned investigatory activity and reduced levels of locomotor habituation that isolation-reared rats displayed in the behavioral pattern monitor. Finally, isolation-reared rats also exhibited sensorimotor gating deficits, reflected by decreased prepulse inhibition of the startle response, consistent with previous studies. We concluded that isolation rearing constitutes a valuable, noninvasive manipulation for modeling schizophrenia-like cognitive deficits and assessing putative therapeutics.
Collapse
|
42
|
Jung SH, Park JM, Moon E, Chung YI, Lee BD, Lee YM, Kim JH, Kim SY, Jeong HJ. Delay in the recovery of normal sleep-wake cycle after disruption of the light-dark cycle in mice: a bipolar disorder-prone animal model? Psychiatry Investig 2014; 11:487-91. [PMID: 25395982 PMCID: PMC4225215 DOI: 10.4306/pi.2014.11.4.487] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/16/2013] [Accepted: 09/30/2013] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Disruption of the circadian rhythm is known as a provoking factor for manic episodes. Individual differences exist in the recovery rate from disruption in the general population. To develop a screening method to detect individuals vulnerable to bipolar disorder, the authors observed the relationship between the recovery of the normal sleep-wake cycle after switching the light-dark (LD) cycle and quinpirole-induced hyperactivity in mice. METHODS Sixteen male mice (age of 5 weeks, weight 28-29 gm) were subjected to a circadian rhythm disruption protocol. Sleep-wake behaviors were checked every 5 min for a total duration of 15 days, i.e., 2 days of baseline observations, 3 days of LD cycle changes, and 10 days of recovery. During the dark cycle on the 16th experimental day, their general locomotor activities were measured in an open field for 120 minutes after an injection of quinpirole (0.5 mg/kg, s.c.). RESULTS The individual differences in the recovery rate of the baseline sleep-wake cycle were noted after 3 days of switching the LD cycle. Fifty percent (n=8) of the mice returned to the baseline cycle within 6 days after normalizing the LD cycle (early recovery group). The locomotor activities of mice that failed to recover within 6 days (delayed recovery group) were significantly higher (mean rank=12.25) than those of the early recovery group (mean rank=4.75, u=62.0, p=0.001, Mann-Whitney U test). CONCLUSION Given that the quinpirole-induced hyperactivity is an animal model of bipolar disorder, our results suggest individuals who have difficulties in recovery from circadian rhythm disruption may be vulnerable to bipolar disorder.
Collapse
Affiliation(s)
- Sun Hwa Jung
- Department of Psychiatry, Pusan National University Hospital, Busan, Republic of Korea
| | - Je-Min Park
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Eunsoo Moon
- Department of Psychiatry, Pusan National University Hospital, Busan, Republic of Korea
| | - Young In Chung
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Byung Dae Lee
- Department of Psychiatry, Pusan National University Hospital, Busan, Republic of Korea
| | - Young Min Lee
- Department of Psychiatry, Pusan National University Hospital, Busan, Republic of Korea
| | - Ji Hoon Kim
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Soo Yeon Kim
- Department of Psychiatry, Pusan National University Hospital, Busan, Republic of Korea
| | - Hee Jeong Jeong
- Department of Psychiatry, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
43
|
Pereira M, Andreatini R, Schwarting RKW, Brenes JC. Amphetamine-induced appetitive 50-kHz calls in rats: a marker of affect in mania? Psychopharmacology (Berl) 2014; 231:2567-77. [PMID: 24414610 DOI: 10.1007/s00213-013-3413-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 12/18/2013] [Indexed: 11/30/2022]
Abstract
RATIONALE Animal models aimed to mimic mania have in common the lack of genuine affective parameters. Although rodent amphetamine-induced hyperlocomotion is a frequently used behavioral model of mania, locomotor activity is a rather unspecific target for developing new pharmacological therapies, and does not necessarily constitute a cardinal symptom in bipolar disorder (BD). Hence, alternative behavioral markers sensitive to stimulants are required. OBJECTIVES Since D-amphetamine induces appetitive 50-kHz ultrasonic vocalizations (USV) in rats, we asked whether established or potential antimanic drugs would inhibit this effect, thereby possibly complementing traditional analysis of locomotor activity. METHODS Amphetamine-treated rats (2.5 mg/kg) were systemically administered with the antimanic drugs lithium (100 mg/kg) and tamoxifen (1 mg/kg). Since protein kinase C (PKC) activity has been implicated in the pathophysiology of bipolar disorder and the biochemical effects of mood stabilizers, the new PKC inhibitor myricitrin (10, 30 mg/kg) was also evaluated. RESULTS We demonstrate for the first time that drugs with known or potential antimanic activity were effective in reversing amphetamine-induced appetitive 50-kHz calls. Treatments particularly normalized amphetamine-induced increases of frequency-modulated calls, a subtype presumably indicative of positive affect in the rat. CONCLUSIONS Our findings suggest that amphetamine-induced 50-kHz calls might constitute a marker for communicating affect that provides a useful model of exaggerated euphoric mood and pressured speech. The antimanic-like effects of the PKC inhibitors tamoxifen and myricitrin support the predictive and etiological validity of both drugs in this model and highlight the role of PKC signaling as a promising target to treat mania and psychosis-related disorders.
Collapse
Affiliation(s)
- Marcela Pereira
- Department of Pharmacology, Federal University of Paraná (UFPR), Curitiba, PR 81540-990, Brazil
| | | | | | | |
Collapse
|
44
|
Stewart AM, Braubach O, Spitsbergen J, Gerlai R, Kalueff AV. Zebrafish models for translational neuroscience research: from tank to bedside. Trends Neurosci 2014; 37:264-78. [PMID: 24726051 DOI: 10.1016/j.tins.2014.02.011] [Citation(s) in RCA: 455] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 01/23/2023]
Abstract
The zebrafish (Danio rerio) is emerging as a new important species for studying mechanisms of brain function and dysfunction. Focusing on selected central nervous system (CNS) disorders (brain cancer, epilepsy, and anxiety) and using them as examples, we discuss the value of zebrafish models in translational neuroscience. We further evaluate the contribution of zebrafish to neuroimaging, circuit level, and drug discovery research. Outlining the role of zebrafish in modeling a wide range of human brain disorders, we also summarize recent applications and existing challenges in this field. Finally, we emphasize the potential of zebrafish models in behavioral phenomics and high-throughput genetic/small molecule screening, which is critical for CNS drug discovery and identifying novel candidate genes.
Collapse
Affiliation(s)
- Adam Michael Stewart
- ZENEREI Institute and the International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Oliver Braubach
- Center for Functional Connectomics, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seoul, 136791, Republic of Korea
| | - Jan Spitsbergen
- Department of Microbiology, Oregon State University, Nash Hall 220 Corvallis, OR 97331, USA
| | - Robert Gerlai
- Department of Psychology, University of Toronto at Mississauga, 3359 Mississauga Road, N Mississauga, Ontario L5L 1C6, Canada
| | - Allan V Kalueff
- ZENEREI Institute and the International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
45
|
Kalueff AV, Nguyen M. Testing anxiolytic drugs in the C57BL/6J mouse strain. J Pharmacol Toxicol Methods 2014; 69:205-7. [DOI: 10.1016/j.vascn.2014.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
46
|
Kirshenbaum G, Burgess C, Déry N, Fahnestock M, Peever J, Roder J. Attenuation of mania-like behavior in Na+,K+-ATPase α3 mutant mice by prospective therapies for bipolar disorder: Melatonin and exercise. Neuroscience 2014; 260:195-204. [DOI: 10.1016/j.neuroscience.2013.12.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/21/2013] [Accepted: 12/04/2013] [Indexed: 01/12/2023]
|
47
|
Arunagiri P, Rajeshwaran K, Shanthakumar J, Balamurugan E. Supplementation of omega-3 fatty acids with aripiprazole and lithium lessens methylphenidate induced manic behavior in Swiss albino mice. PHARMANUTRITION 2014. [DOI: 10.1016/j.phanu.2013.11.128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
48
|
Henry BL, Minassian A, Patt V, Hua J, Young JW, Geyer MA, Perry W. Inhibitory deficits in euthymic bipolar disorder patients assessed in the human behavioral pattern monitor. J Affect Disord 2013; 150:948-54. [PMID: 23759280 PMCID: PMC3759601 DOI: 10.1016/j.jad.2013.05.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 05/03/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Bipolar disorder (BD) is associated with inhibitory deficits characterized by a reduced ability to control inappropriate actions or thoughts. While aspects of inhibition such as exaggerated novelty-seeking and perseveration are quantified in rodent exploration of novel environments, similar models are rarely applied in humans. The human Behavioral Pattern Monitor (hBPM), a cross-species exploratory paradigm, has identified a pattern of impaired inhibitory function in manic BD participants, but this phenotype has not been examined across different BD phases. The objective of this study was to determine if euthymic BD individuals demonstrate inhibitory deficits in the hBPM, supporting disinhibition as an endophenotype for the disorder. METHODS 25 euthymic BD outpatients and 51 healthy comparison subjects were assessed in the hBPM, where activity was recorded by a concealed videocamera and an ambulatory monitoring sensor. RESULTS Euthymic BD individuals, similar to manic subjects, demonstrated increased motor activity, greater interaction with novel objects, and more frequent perseverative behavior relative to comparison participants. The quantity of locomotion was also reduced in BD individuals treated with mood stabilizers compared to other patients. LIMITATIONS Low sample size for treatment subgroups limits the evaluation of specific medication regimens. CONCLUSIONS Our results suggest that BD is distinguished by both trait- and state-dependent inhibitory deficits optimally assessed with sophisticated multivariate measures. These data support the use of the hBPM as a tool to elucidate the effects of BD across various illness states, facilitate the development of BD animal models, and advance our understanding of the neurobiology underlying the disorder.
Collapse
Affiliation(s)
- Brook L. Henry
- University of California San Diego, Department of Psychiatry, La Jolla, CA
| | - Arpi Minassian
- University of California San Diego, Department of Psychiatry, La Jolla, CA,Research Service, VA San Diego Healthcare System, San Diego, CA
| | - Virginie Patt
- University of California San Diego, Department of Psychiatry, La Jolla, CA
| | - Jessica Hua
- University of California San Diego, Department of Psychiatry, La Jolla, CA
| | - Jared W. Young
- University of California San Diego, Department of Psychiatry, La Jolla, CA
| | - Mark A. Geyer
- University of California San Diego, Department of Psychiatry, La Jolla, CA,Research Service, VA San Diego Healthcare System, San Diego, CA
| | - William Perry
- University of California San Diego, Department of Psychiatry, La Jolla, CA
| |
Collapse
|
49
|
Abstract
Bipolar disorder (BD) mania is a psychiatric disorder with multifaceted symptoms. Development of targeted treatments for BD mania may benefit from animal models that mimic multiple symptoms, as opposed to hyperactivity alone. Using the reverse-translated multivariate exploratory paradigm, the behavioural pattern monitor (BPM), we reported that patients with BD mania exhibit hyperactivity as well as increased specific exploration and more linear movements through space. This abnormal profile is also observed in mice with reduced function of the dopamine transporter (DAT) through either constitutive genetic [knockdown (KD)] or acute pharmacological (GBR12909) means. Here, we assessed the pharmacological predictive validity of these models by administering the BD-treatment valproic acid (VPA) for 28 d. After 1.5% VPA- or regular-chow treatment for 28 d, C57BL/6J mice received GBR12909 (9 mg/kg) or saline and were tested in the BPM. Similarly, DAT KD and wild type (WT) littermates were treated with VPA-chow and tested in the BPM. GBR12909-treated and DAT KD mice on regular chow were hyperactive, exhibited increased specific exploration and moved in straighter patterns compared to saline-treated and WT mice respectively. Chronic 1.5% VPA-chow treatment resulted in therapeutic concentrations of VPA and ameliorated hyperactivity in both models, while specific exploration and behavioural organization remained unaffected. Hence, the mania-like profile of mice with reduced functional DAT was partially attenuated by chronic VPA treatment, consistent with the incomplete symptomatic effect of VPA treatment in BD patients. Both DAT models may help to identify therapeutics that impact the full spectrum of BD mania.
Collapse
|
50
|
van Enkhuizen J, Minassian A, Young JW. Further evidence for ClockΔ19 mice as a model for bipolar disorder mania using cross-species tests of exploration and sensorimotor gating. Behav Brain Res 2013; 249:44-54. [PMID: 23623885 DOI: 10.1016/j.bbr.2013.04.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/12/2013] [Accepted: 04/16/2013] [Indexed: 02/07/2023]
Abstract
Bipolar disorder (BD) is a pervasive neuropsychiatric disorder characterized by episodes of mania and depression. The switch between mania and depression may reflect seasonal changes and certainly can be affected by alterations in sleep and circadian control. The circadian locomotor output cycles kaput (CLOCK) protein is a key component of the cellular circadian clock. Mutation of the Clock gene encoding this protein in ClockΔ19 mutant mice leads to behavioral abnormalities reminiscent of BD mania. To date, however, these mice have not been assessed in behavioral paradigms that have cross-species translational validity. In the present studies of ClockΔ19 and wildtype (WT) littermate mice, we quantified exploratory behavior and sensorimotor gating, which are abnormal in BD manic patients. We also examined the saccharin preference of these mice and their circadian control in different photoperiods. ClockΔ19 mice exhibited behavioral alterations that are consistent with BD manic patients tested in comparable tasks, including hyperactivity, increased specific exploration, and reduced sensorimotor gating. Moreover, compared to WT mice, ClockΔ19 mice exhibited a greater preference for sweetened solutions and greater sensitivity to altered photoperiod. In contrast with BD manic patients however, ClockΔ19 mice exhibited more circumscribed movements during exploration. Future studies will extend the characterization of these mice in measures with cross-species translational relevance to human testing.
Collapse
Affiliation(s)
- Jordy van Enkhuizen
- Department of Psychiatry, University of California, San Diego (UCSD), 9500 Gilman Drive MC 0804, La Jolla, CA 92093-0804, United States
| | | | | |
Collapse
|