301
|
Sun X, Zhang Y, Li X, Liu X, Qin C. Early-Life Neglect Alters Emotional and Cognitive Behavior in a Sex-Dependent Manner and Reduces Glutamatergic Neuronal Excitability in the Prefrontal Cortex. Front Psychiatry 2020; 11:572224. [PMID: 33574771 PMCID: PMC7870800 DOI: 10.3389/fpsyt.2020.572224] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 12/07/2020] [Indexed: 01/19/2023] Open
Abstract
Early-life neglect in critical developmental periods has been associated with emotional and cognitive consequences. Maternal separation (MS) has been commonly used as a rodent model to identify the developmental effects of child neglect. However, reports have shown considerable variability in behavioral results from MS studies in both mice and rats. Difficulties in developing reliable child neglect models have impeded advances in identifying the effects of early-life stress. Accumulating evidence shows that neuronal intrinsic excitability plays an important role in information processing and storage in the brain. The prefrontal cortex (PFC) integrates information from many cortical and subcortical structures. No studies to date have examined the impact of early-life stress on glutamatergic neuronal excitability in the PFC. This study aimed to develop a reliable child neglect rat model and observe glutamatergic neuronal excitability in the PFC. An MS with early weaning (MSEW) rat model was developed. Rats were separated from the dam for 4 h per day on postnatal days (PNDs) 2-5 and for 8 h per day on PNDs 6-16 and then weaned on PND 17. A battery of behavioral tests was used to assess anxiety-like behavior, coping behavior, working memory, spatial reference memory, and fear memory. The action potentials (APs) of glutamatergic neuronal membranes were recorded. MSEW resulted in anxiety-like behavior, a passive coping strategy and increased fear memory in male rats and decreased locomotor activity in both sexes. MSEW slightly impaired working memory during non-stressful situations in female rats but did not change spatial reference memory or associative learning under stressful circumstances in either sex. MSEW reduced the number of glutamatergic neuron APs in male rats. Our findings showed that MS with early weaning induced anxiety-like behavior in male rats. The reduced glutamatergic neuronal excitability may be associated with the emotional alteration induced by MSEW in male rats. In addition, MSEW induced adaptive modification, which depended on a non-stressful context.
Collapse
Affiliation(s)
- Xiuping Sun
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yu Zhang
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Xianglei Li
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Xinmin Liu
- Peking Union Medical College, Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Beijing, China
| | - Chuan Qin
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| |
Collapse
|
302
|
Mirbozorgi SA, Jia Y, Zhang P, Ghovanloo M. Toward a High-Throughput Wireless Smart Arena for Behavioral Experiments on Small Animals. IEEE Trans Biomed Eng 2019; 67:2359-2369. [PMID: 31870973 DOI: 10.1109/tbme.2019.2961297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This work presents a high-throughput and scalable wirelessly-powered smart arena for behavioral experiments made of multiple EnerCage Homecage (HC) systems, operating in parallel in a way that they can fit in standard racks that are commonly used in animal facilities. The proposed system, which is referred to as the multi-EnerCage-HC (mEHC), increases the volume of data that can be collected from more animal subjects, while lowering the cost and duration of experiments as well as stress-induced bias by minimizing the involvement of human operators. Thus improving the quality, reproducibility, and statistical power of experiment outcomes, while saving precious lab space. The system is equipped with an auto-tuning mechanism to compensate for the resonance frequency shifts caused by the dynamic nature of the mutual inductance between adjacent homecages. A functional prototype of the mEHC system has been implemented with 7 units and analyzed for theoretical design considerations that would minimize the effects of interference and resonance frequency bifurcation. Experiment results demonstrate robust wireless power and data transmissions capabilities of this system within the noisy lab environment.
Collapse
|
303
|
Wood T, Nance E. Disease-directed engineering for physiology-driven treatment interventions in neurological disorders. APL Bioeng 2019; 3:040901. [PMID: 31673672 PMCID: PMC6811362 DOI: 10.1063/1.5117299] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
Neurological disease is killing us. While there have long been attempts to develop therapies for both acute and chronic neurological diseases, no current treatments are curative. Additionally, therapeutic development for neurological disease takes 15 years and often costs several billion dollars. More than 96% of these therapies will fail in late stage clinical trials. Engineering novel treatment interventions for neurological disease can improve outcomes and quality of life for millions; however, therapeutics should be designed with the underlying physiology and pathology in mind. In this perspective, we aim to unpack the importance of, and need to understand, the physiology of neurological disease. We first dive into the normal physiological considerations that should guide experimental design, and then assess the pathophysiological factors of acute and chronic neurological disease that should direct treatment design. We provide an analysis of a nanobased therapeutic intervention that proved successful in translation due to incorporation of physiology at all stages of the research process. We also provide an opinion on the importance of keeping a high-level view to designing and administering treatment interventions. Finally, we close with an implementation strategy for applying a disease-directed engineering approach. Our assessment encourages embracing the complexity of neurological disease, as well as increasing efforts to provide system-level thinking in our development of therapeutics for neurological disease.
Collapse
|
304
|
Roig-Puiggros S, Vigouroux RJ, Beckman D, Bocai NI, Chiou B, Davimes J, Gomez G, Grassi S, Hoque A, Karikari TK, Kiffer F, Lopez M, Lunghi G, Mazengenya P, Meier S, Olguín-Albuerne M, Oliveira MM, Paraíso-Luna J, Pradhan J, Radiske A, Ramos-Hryb AB, Ribeiro MC, Schellino R, Selles MC, Singh S, Theotokis P, Chédotal A. Construction and reconstruction of brain circuits: normal and pathological axon guidance. J Neurochem 2019; 153:10-32. [PMID: 31630412 DOI: 10.1111/jnc.14900] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
Perception of our environment entirely depends on the close interaction between the central and peripheral nervous system. In order to communicate each other, both systems must develop in parallel and in coordination. During development, axonal projections from the CNS as well as the PNS must extend over large distances to reach their appropriate target cells. To do so, they read and follow a series of axon guidance molecules. Interestingly, while these molecules play critical roles in guiding developing axons, they have also been shown to be critical in other major neurodevelopmental processes, such as the migration of cortical progenitors. Currently, a major hurdle for brain repair after injury or neurodegeneration is the absence of axonal regeneration in the mammalian CNS. By contrasts, PNS axons can regenerate. Many hypotheses have been put forward to explain this paradox but recent studies suggest that hacking neurodevelopmental mechanisms may be the key to promote CNS regeneration. Here we provide a seminar report written by trainees attending the second Flagship school held in Alpbach, Austria in September 2018 organized by the International Society for Neurochemistry (ISN) together with the Journal of Neurochemistry (JCN). This advanced school has brought together leaders in the fields of neurodevelopment and regeneration in order to discuss major keystones and future challenges in these respective fields.
Collapse
Affiliation(s)
| | - Robin J Vigouroux
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Danielle Beckman
- California National Primate Research Center, UC Davis, Davis, California, USA
| | - Nadia I Bocai
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brian Chiou
- Department of Pediatrics, University of California - San Francisco, San Francisco, California, USA
| | - Joshua Davimes
- Faculty of Health Sciences School of Anatomical Sciences, University of the Witwatersrand, Parktown Johannesburg, South Africa
| | - Gimena Gomez
- Laboratorio de Parkinson Experimental, Instituto de Investigaciones Farmacológicas (ININFA-CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Ashfaqul Hoque
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,School of Life Sciences, University of Warwick, Coventry, UK.,Midlands Integrative Biosciences Training Partnership, University of Warwick, Coventry, UK
| | - Frederico Kiffer
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Mary Lopez
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicin, University of Milano, Segrate, Italy
| | - Pedzisai Mazengenya
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sonja Meier
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mauricio M Oliveira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juan Paraíso-Luna
- Ramón y Cajal Institute of Health Research (IRYCIS), Department of Biochemistry and Molecular Biology and University Research Institute in Neurochemistry (IUIN), Complutense University, Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jonu Pradhan
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Andressa Radiske
- Memory Research Laboratory, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ana Belén Ramos-Hryb
- Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina.,Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Mayara C Ribeiro
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York, USA
| | - Roberta Schellino
- Neuroscience Department "Rita Levi-Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy
| | - Maria Clara Selles
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Shripriya Singh
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Paschalis Theotokis
- Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Macedonia, Greece
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
305
|
Doulberis M, Papaefthymiou A, Polyzos SA, Katsinelos P, Grigoriadis N, Srivastava DS, Kountouras J. Rodent models of obesity. MINERVA ENDOCRINOL 2019; 45:243-263. [PMID: 31738033 DOI: 10.23736/s0391-1977.19.03058-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Obese or overweight people exceed one-third of the global population and obesity along with diabetes mellitus consist basic components of metabolic syndrome, both of which are known cardio-cerebrovascular risk factors with detrimental consequences. These data signify the pandemic character of obesity and the necessity for effective treatments. Substantial advances have been accomplished in preclinical research of obesity by using animal models, which mimic the human disease. In particular, rodent models have been widely used for many decades with success for the elucidation of the pathophysiology of obesity, since they share physiological and genetic components with humans and appear advantageous in their husbandry. The most representative rodents include the laboratory mouse and rat. Within this review, we attempted to consolidate the most widely used mice and rat models of obesity and highlight their strengths as well as weaknesses in a critical way. Our aim was to bridge the gap between laboratory facilities and patient's bed and help the researcher find the appropriate animal model for his/her obesity research. This tactful selection of the appropriate model of obesity may offer more translational derived results. In this regard, we included, the main diet induced models, the chemical/mechanical ones, as well as a selection of monogenic or polygenic models.
Collapse
Affiliation(s)
- Michael Doulberis
- Department of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland - .,Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece -
| | | | | | - Panagiotis Katsinelos
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- First Department of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - David S Srivastava
- Second Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Jannis Kountouras
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
306
|
Planchez B, Surget A, Belzung C. Animal models of major depression: drawbacks and challenges. J Neural Transm (Vienna) 2019; 126:1383-1408. [PMID: 31584111 PMCID: PMC6815270 DOI: 10.1007/s00702-019-02084-y] [Citation(s) in RCA: 239] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Abstract
Major depression is a leading contributor to the global burden of disease. This situation is mainly related to the chronicity and/or recurrence of the disorder, and to poor response to antidepressant therapy. Progress in this area requires valid animal models. Current models are based either on manipulating the environment to which rodents are exposed (during the developmental period or adulthood) or biological underpinnings (i.e. gene deletion or overexpression of candidate genes, targeted lesions of brain areas, optogenetic control of specific neuronal populations, etc.). These manipulations can alter specific behavioural and biological outcomes that can be related to different symptomatic and pathophysiological dimensions of major depression. However, animal models of major depression display substantial shortcomings that contribute to the lack of innovative pharmacological approaches in recent decades and which hamper our capabilities to investigate treatment-resistant depression. Here, we discuss the validity of these models, review putative models of treatment-resistant depression, major depression subtypes and recurrent depression. Furthermore, we identify future challenges regarding new paradigms such as those proposing dimensional rather than categorical approaches to depression.
Collapse
Affiliation(s)
| | | | - Catherine Belzung
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
- UMR 1253, iBrain, UFR Sciences et Techniques, Parc Grandmont, 37200, Tours, France.
| |
Collapse
|
307
|
Scheyer AF, Melis M, Trezza V, Manzoni OJJ. Consequences of Perinatal Cannabis Exposure. Trends Neurosci 2019; 42:871-884. [PMID: 31604585 DOI: 10.1016/j.tins.2019.08.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/10/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
Abstract
Cannabis exposure during the perinatal period results in varied and significant consequences in affected offspring. The prevalence of detrimental outcomes of perinatal cannabis exposure is likely to increase in tandem with the broadening of legalization and acceptance of the drug. As such, it is crucial to highlight the immediate and protracted consequences of cannabis exposure on pre- and postnatal development. Here, we identify lasting changes in neurons' learning flexibility (synaptic plasticity) and epigenetic misregulation in animal models of perinatal cannabinoid exposure (using synthetic cannabinoids or active components of the cannabis plant), in addition to significant alterations in social behavior and executive functions. These findings are supported by epidemiological data indicating similar behavioral outcomes throughout life in human offspring exposed to cannabis during pregnancy. Further, we indicate important lingering questions regarding accurate modeling of perinatal cannabis exposure as well as the need for sex- and age-dependent outcome measures in future studies.
Collapse
Affiliation(s)
- Andrew F Scheyer
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Provence, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Aix-Marseille University, Provence, France/Indiana University, Bloomington, IN, USA
| | - Miriam Melis
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; National Institute of Neuroscience, Cagliari, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Olivier J J Manzoni
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Provence, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Aix-Marseille University, Provence, France/Indiana University, Bloomington, IN, USA.
| |
Collapse
|
308
|
Zhang P, Miller EB, Manna SK, Meleppat RK, Pugh EN, Zawadzki RJ. Temporal speckle-averaging of optical coherence tomography volumes for in-vivo cellular resolution neuronal and vascular retinal imaging. NEUROPHOTONICS 2019; 6:041105. [PMID: 31528657 PMCID: PMC6732665 DOI: 10.1117/1.nph.6.4.041105] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/31/2019] [Indexed: 05/08/2023]
Abstract
It has been recently demonstrated that structures corresponding to the cell bodies of highly transparent cells in the retinal ganglion cell layer could be visualized noninvasively in the living human eye by optical coherence tomography (OCT) via temporal averaging. Inspired by this development, we explored the application of volumetric temporal averaging in mice, which are important models for studying human retinal diseases and therapeutic interventions. A general framework of temporal speckle-averaging (TSA) of OCT and optical coherence tomography angiography (OCTA) is presented and applied to mouse retinal volumetric data. Based on the image analysis, the eyes of mice under anesthesia exhibit only minor motions, corresponding to lateral displacements of a few micrometers and rotations of a fraction of 1 deg. Moreover, due to reduced eye movements under anesthesia, there is a negligible amount of motion artifacts within the volumes that need to be corrected to achieve volume coregistration. In addition, the relatively good optical quality of the mouse ocular media allows for cellular-resolution imaging without adaptive optics (AO), greatly simplifying the experimental system, making the proposed framework feasible for large studies. The TSA OCT and TSA OCTA results provide rich information about new structures previously not visualized in living mice with non-AO-OCT. The mechanism of TSA relies on improving signal-to-noise ratio as well as efficient suppression of speckle contrast due to temporal decorrelation of the speckle patterns, enabling full utilization of the high volumetric resolution offered by OCT and OCTA.
Collapse
Affiliation(s)
- Pengfei Zhang
- University of California Davis, Department of Cell Biology and Human Anatomy, UC Davis Eye-Pod Small Animal Ocular Imaging Laboratory, Davis, California, United States
| | - Eric B. Miller
- University of California Davis, Center for Neuroscience, Davis, California, United States
| | - Suman K. Manna
- University of California Davis, Department of Cell Biology and Human Anatomy, UC Davis Eye-Pod Small Animal Ocular Imaging Laboratory, Davis, California, United States
| | - Ratheesh K. Meleppat
- University of California Davis, Department of Cell Biology and Human Anatomy, UC Davis Eye-Pod Small Animal Ocular Imaging Laboratory, Davis, California, United States
| | - Edward N. Pugh
- University of California Davis, Department of Cell Biology and Human Anatomy, UC Davis Eye-Pod Small Animal Ocular Imaging Laboratory, Davis, California, United States
- University of California Davis, Department of Ophthalmology and Vision Science, Vision Science and Advanced Retinal Imaging Laboratory, Sacramento, California, United States
| | - Robert J. Zawadzki
- University of California Davis, Department of Cell Biology and Human Anatomy, UC Davis Eye-Pod Small Animal Ocular Imaging Laboratory, Davis, California, United States
- University of California Davis, Department of Ophthalmology and Vision Science, Vision Science and Advanced Retinal Imaging Laboratory, Sacramento, California, United States
- University of California Davis, UC Davis Eye Center, Department of Ophthalmology and Vision Science, Sacramento, California, United States
- Address all correspondence to Robert J. Zawadzki, E-mail:
| |
Collapse
|
309
|
Graham BA, Hughes DI. Rewards, perils and pitfalls of untangling spinal pain circuits. CURRENT OPINION IN PHYSIOLOGY 2019. [DOI: 10.1016/j.cophys.2019.04.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
310
|
Huang C, Mazdeyasna S, Chen L, Abu Jawdeh EG, Bada HS, Saatman KE, Chen L, Yu G. Noninvasive noncontact speckle contrast diffuse correlation tomography of cerebral blood flow in rats. Neuroimage 2019; 198:160-169. [DOI: 10.1016/j.neuroimage.2019.05.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/05/2023] Open
|
311
|
Avoiding Beach’s Boojum Effect: Enhancing bench to bedside translation with field to laboratory considerations in optimal animal models. Neurosci Biobehav Rev 2019; 104:191-196. [DOI: 10.1016/j.neubiorev.2019.06.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/15/2019] [Accepted: 06/24/2019] [Indexed: 11/23/2022]
|
312
|
Abstract
Animal models are indispensable tools for Alzheimer disease (AD) research. Over the course of more than two decades, an increasing number of complementary rodent models has been generated. These models have facilitated testing hypotheses about the aetiology and progression of AD, dissecting the associated pathomechanisms and validating therapeutic interventions, thereby providing guidance for the design of human clinical trials. However, the lack of success in translating rodent data into therapeutic outcomes may challenge the validity of the current models. This Review critically evaluates the genetic and non-genetic strategies used in AD modelling, discussing their strengths and limitations, as well as new opportunities for the development of better models for the disease.
Collapse
|
313
|
Mäki-Marttunen T, Kaufmann T, Elvsåshagen T, Devor A, Djurovic S, Westlye LT, Linne ML, Rietschel M, Schubert D, Borgwardt S, Efrim-Budisteanu M, Bettella F, Halnes G, Hagen E, Næss S, Ness TV, Moberget T, Metzner C, Edwards AG, Fyhn M, Dale AM, Einevoll GT, Andreassen OA. Biophysical Psychiatry-How Computational Neuroscience Can Help to Understand the Complex Mechanisms of Mental Disorders. Front Psychiatry 2019; 10:534. [PMID: 31440172 PMCID: PMC6691488 DOI: 10.3389/fpsyt.2019.00534] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
The brain is the most complex of human organs, and the pathophysiology underlying abnormal brain function in psychiatric disorders is largely unknown. Despite the rapid development of diagnostic tools and treatments in most areas of medicine, our understanding of mental disorders and their treatment has made limited progress during the last decades. While recent advances in genetics and neuroscience have a large potential, the complexity and multidimensionality of the brain processes hinder the discovery of disease mechanisms that would link genetic findings to clinical symptoms and behavior. This applies also to schizophrenia, for which genome-wide association studies have identified a large number of genetic risk loci, spanning hundreds of genes with diverse functionalities. Importantly, the multitude of the associated variants and their prevalence in the healthy population limit the potential of a reductionist functional genetics approach as a stand-alone solution to discover the disease pathology. In this review, we outline the key concepts of a "biophysical psychiatry," an approach that employs large-scale mechanistic, biophysics-founded computational modelling to increase transdisciplinary understanding of the pathophysiology and strive toward robust predictions. We discuss recent scientific advances that allow a synthesis of previously disparate fields of psychiatry, neurophysiology, functional genomics, and computational modelling to tackle open questions regarding the pathophysiology of heritable mental disorders. We argue that the complexity of the increasing amount of genetic data exceeds the capabilities of classical experimental assays and requires computational approaches. Biophysical psychiatry, based on modelling diseased brain networks using existing and future knowledge of basic genetic, biochemical, and functional properties on a single neuron to a microcircuit level, may allow a leap forward in deriving interpretable biomarkers and move the field toward novel treatment options.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Anna Devor
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Dirk Schubert
- Cognitive Neuroscience Department, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Stefan Borgwardt
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Magdalena Efrim-Budisteanu
- Prof. Dr. Alex. Obregia Clinical Hospital of Psychiatry, Bucharest, Romania
- Victor Babes National Institute of Pathology, Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Francesco Bettella
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Geir Halnes
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Espen Hagen
- Department of Physics, University of Oslo, Oslo, Norway
| | - Solveig Næss
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Torbjørn V. Ness
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Torgeir Moberget
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christoph Metzner
- Centre for Computer Science and Informatics Research, University of Hertfordshire, Hatfield, United Kingdom
- Institute of Software Engineering and Theoretical Computer Science, Technische Universität zu Berlin, Berlin, Germany
| | - Andrew G. Edwards
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Anders M. Dale
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Gaute T. Einevoll
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
314
|
Sex still matters: has the prevalence of male-only studies of drug effects on rodent behaviour changed during the past decade? Behav Pharmacol 2019; 30:95-99. [PMID: 29847339 DOI: 10.1097/fbp.0000000000000410] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
During the past 10 years, for a number of biomedical disciplines, including behavioural pharmacology, there have been appeals to include both sexes in animal studies of processes that are not sex specific. In 2007, a survey of experimental studies of drug or other chemical effects on rodent behaviour, published in five prominent journals over 20 months (February 2005 to September 2006, inclusive), revealed that 85% of these conducted with rats and 78% of these conducted with mice involved males only. This was in spite of the evidence of sex differences in responsiveness to an increasing number of compounds. To see if the situation has improved, the survey was repeated with the same journals for a comparable period namely, February 2016 to September 2017 (inclusive). Even though there have been repeated appeals for biomedical research that is not sex specific to involve both sexes, it was apparent that little has changed since 2005-2006, as 82% of rat and 75% of mouse studies were again conducted with males only. However, there was an increase in studies with mice, which may be owing to a greater interest in genetic factors. The male-only situation could be rectified by appropriate funding agencies and journals that publish behavioural pharmacological research insisting that both sexes must be included in research that is not sex specific along with valid scientific justification for single-sex studies, as now typifies some other disciplines.
Collapse
|
315
|
Morrison TJ, Sefton E, Marquez-Chin M, Popovic MR, Morshead CM, Naguib HE. A 3D Printed Device for Low Cost Neural Stimulation in Mice. Front Neurosci 2019; 13:784. [PMID: 31417347 PMCID: PMC6682623 DOI: 10.3389/fnins.2019.00784] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/12/2019] [Indexed: 12/13/2022] Open
Abstract
Electrical stimulation of the brain through the implantation of electrodes is an effective treatment for certain diseases and the focus of a large body of research investigating new cell mechanisms, neurological phenomena, and treatments. Electrode devices developed for stimulation in rodents vary widely in size, cost, and functionality, with the majority of recent studies presenting complex, multi-functional designs. While some experiments require these added features, others are in greater need of reliable, low cost, and readily available devices that will allow surgeries to be scheduled and completed without delay. In this work, we utilize 3D printing and common electrical hardware to produce an effective 2-channel stimulation device that meets these requirements. Our stimulation electrode has not failed in over 60 consecutive surgeries, costs less than $1 USD, and can be assembled in less than 20 min. 3D printing minimizes the amount of material used in manufacturing the device and enables one to match the curvature of the connector’s base with the curvature of the mouse skull, producing an ultra-lightweight, low size device with improved adhesion to the mouse skull. The range of the stimulation parameters used with the proposed device was: pulse amplitude 1–200 μA, pulse duration 50–500 μs and pulse frequency 1–285 Hz.
Collapse
Affiliation(s)
- Taylor J Morrison
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Elana Sefton
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Melissa Marquez-Chin
- Department of Engineering, Universidad Iberoamericana, Mexico City, Mexico.,KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Milos R Popovic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Cindi M Morshead
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Hani E Naguib
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Department of Materials Science & Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
316
|
Zaytseva AS, Volodin IA, Ilchenko OG, Volodina EV. Ultrasonic vocalization of pup and adult fat-tailed gerbils (Pachyuromys duprasi). PLoS One 2019; 14:e0219749. [PMID: 31356642 PMCID: PMC6663002 DOI: 10.1371/journal.pone.0219749] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 07/01/2019] [Indexed: 12/03/2022] Open
Abstract
Ultrasonic vocalizations (USVs) of laboratory rodents indicate animal emotional arousal and may serve as models of human disorders. We analysed spectrographically USV calls of pup and adult fat-tailed gerbils Pachyuromys duprasi during 420-s tests, including isolation, touch and handling. Based on combination of six different USV syllable contour shapes and six different note compositions, we classified 782 USV syllables of 24 pups aged 5-10 days to 18 types and 232 syllables of 7 adults to 24 types. Pups and adults shared 16 of these 26 USV types. Percentages of USV syllables with certain contour shapes differed between pups and adults. The contour shape and note composition significantly affected most acoustic variables of USV syllables in either pups or adults. The 1-note USV syllables were most common in either pups or adults. Pup USV syllables were overall longer and higher-frequency than adult ones, reminiscent of the USV ontogenetic pathway of bats and distinctive to rats and mice. We discuss that the USV syllable types of fat-tailed gerbils were generally similar in contour shapes and note compositions with USV syllable types of mice and rats, what means that software developed for automated classifying of mice ultrasound might be easily adapted or re-tuned to gerbil USV calls. However, using fat-tailed gerbils as model for biomedical research including control of USV vocalization is only possible since 6th day of pup life, because of the delayed emergence of USV calls in ontogeny of this species.
Collapse
Affiliation(s)
- Alexandra S. Zaytseva
- Department of Vertebrate Zoology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
- Scientific Research Department, Moscow Zoo, Moscow, Russia
| | - Ilya A. Volodin
- Department of Vertebrate Zoology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
- Scientific Research Department, Moscow Zoo, Moscow, Russia
| | | | | |
Collapse
|
317
|
Scheimann JR, Moloney RD, Mahbod P, Morano RL, Fitzgerald M, Hoskins O, Packard BA, Cotella EM, Hu YC, Herman JP. Conditional deletion of glucocorticoid receptors in rat brain results in sex-specific deficits in fear and coping behaviors. eLife 2019; 8:44672. [PMID: 31329100 PMCID: PMC6645713 DOI: 10.7554/elife.44672] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 07/03/2019] [Indexed: 01/16/2023] Open
Abstract
Glucocorticoid receptors (GR) have diverse functions relevant to maintenance of homeostasis and adaptation to environmental challenges. Understanding the importance of tissue-specific GR function in physiology and behavior has been hampered by near-ubiquitous localization in brain and body. Here we use CRISPR/Cas9 gene editing to create a conditional GR knockdown in Sprague Dawley rats. To test the impact of cell- and region-specific GR knockdown on physiology and behavior, we targeted GR knockdown to output neurons of the prelimbic cortex. Prelimbic knockdown of GR in females caused deficits in acquisition and extinction of fear memory during auditory fear conditioning, whereas males exhibited enhanced active-coping behavior during forced swim. Our data support the utility of this conditional knockdown rat to afford high-precision knockdown of GR across a variety of contexts, ranging from neuronal depletion to circuit-wide manipulations, leveraging the behavioral tractability and enhanced brain size of the rat as a model organism.
Collapse
Affiliation(s)
- Jessie R Scheimann
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Rachel D Moloney
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Parinaz Mahbod
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Rachel L Morano
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Maureen Fitzgerald
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Olivia Hoskins
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Benjamin A Packard
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Evelin M Cotella
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, United States.,Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - James P Herman
- Department Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, United States
| |
Collapse
|
318
|
Duggal P, Mehan S. Neuroprotective Approach of Anti-Cancer Microtubule Stabilizers Against Tauopathy Associated Dementia: Current Status of Clinical and Preclinical Findings. J Alzheimers Dis Rep 2019; 3:179-218. [PMID: 31435618 PMCID: PMC6700530 DOI: 10.3233/adr-190125] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuronal microtubule (MT) tau protein provides cytoskeleton to neuronal cells and plays a vital role including maintenance of cell shape, intracellular transport, and cell division. Tau hyperphosphorylation mediates MT destabilization resulting in axonopathy and neurotransmitter deficit, and ultimately causing Alzheimer’s disease (AD), a dementing disorder affecting vast geriatric populations worldwide, characterized by the existence of extracellular amyloid plaques and intracellular neurofibrillary tangles in a hyperphosphorylated state. Pre-clinically, streptozotocin stereotaxically mimics the behavioral and biochemical alterations similar to AD associated with tau pathology resulting in MT assembly defects, which proceed neuropathological cascades. Accessible interventions like cholinesterase inhibitors and NMDA antagonist clinically provides only symptomatic relief. Involvement of microtubule stabilizers (MTS) prevents tauopathy particularly by targeting MT oriented cytoskeleton and promotes polymerization of tubulin protein. Multiple in vitro and in vivo research studies have shown that MTS can hold substantial potential for the treatment of AD-related tauopathy dementias through restoration of tau function and axonal transport. Moreover, anti-cancer taxane derivatives and epothiolones may have potential to ameliorate MT destabilization and prevent the neuronal structural and functional alterations associated with tauopathies. Therefore, this current review strictly focuses on exploration of various clinical and pre-clinical features available for AD to understand the neuropathological mechanisms as well as introduce pharmacological interventions associated with MT stabilization. MTS from diverse natural sources continue to be of value in the treatment of cancer, suggesting that these agents have potential to be of interest in the treatment of AD-related tauopathy dementia in the future.
Collapse
Affiliation(s)
- Pallavi Duggal
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
319
|
Redecker TM, Kisko TM, Schwarting RK, Wöhr M. Effects of Cacna1c haploinsufficiency on social interaction behavior and 50-kHz ultrasonic vocalizations in adult female rats. Behav Brain Res 2019; 367:35-52. [DOI: 10.1016/j.bbr.2019.03.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/03/2019] [Accepted: 03/15/2019] [Indexed: 01/28/2023]
|
320
|
Health Benefits of Endurance Training: Implications of the Brain-Derived Neurotrophic Factor-A Systematic Review. Neural Plast 2019; 2019:5413067. [PMID: 31341469 PMCID: PMC6613032 DOI: 10.1155/2019/5413067] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/07/2019] [Accepted: 02/24/2019] [Indexed: 02/06/2023] Open
Abstract
This article presents a concept that wide expression of brain-derived neurotrophic factor (BDNF) and its receptors (TrkB) in the nervous tissue, evoked by regular endurance training (ET), can cause numerous motor and metabolic adaptations, which are beneficial for human health. The relationships between the training-evoked increase of endogenous BDNF and molecular and/or physiological adaptations in the nervous structures controlling both motor performance and homeostasis of the whole organism have been presented. Due to a very wide range of plastic changes that ET has exerted on various systems of the body, the improvement of motor skills and counteraction of the development of civilization diseases resulting from the posttraining increase of BDNF/TrkB levels have been discussed, as important for people, who undertake ET. Thus, this report presents the influence of endurance exercises on the (1) transformation of motoneuron properties, which are a final element of the motor pathways, (2) reduction of motor deficits evoked by Parkinson disease, and (3) prevention of the metabolic syndrome (MetS). This review suggests that the increase of posttraining levels of BDNF and its TrkB receptors causes simultaneous changes in the activity of the spinal cord, the substantia nigra, and the hypothalamic nuclei neurons, which are responsible for the alteration of the functional properties of motoneurons innervating the skeletal muscles, for the enhancement of dopamine release in the brain, and for the modulation of hormone levels involved in regulating the metabolic processes, responsively. Finally, training-evoked increase of the BDNF/TrkB leads to a change in a manner of regulation of skeletal muscles, causes a reduction of motor deficits observed in the Parkinson disease, and lowers weight, glucose level, and blood pressure, which accompany the MetS. Therefore, BDNF seems to be the molecular factor of pleiotropic activity, important in the modulation processes, underlying adaptations, which result from ET.
Collapse
|
321
|
Dingle A, Zeng W, Ness JP, Albano N, Minor RL, Feldman C, Austin M, Brodnick SK, Shulzhenko N, Sanchez R, Lake WB, Williams JC, Poore SO, Suminski AJ. Strategies for interfacing with the trigeminal nerves in rodents for bioelectric medicine. J Neurosci Methods 2019; 324:108321. [PMID: 31229585 DOI: 10.1016/j.jneumeth.2019.108321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/05/2019] [Accepted: 06/19/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Bioelectric medicine seeks to modulate neural activity via targeted electrical stimulation to treat disease. Recent clinical evidence supports trigeminal nerve stimulation as a bioelectric treatment for several neurological disorders; however, the mechanisms of trigeminal nerve stimulation and potential side effects remain largely unknown. The goal of this study is to optimize the methodology and reproducibility of neural interface implantation for mechanistic studies in rodents. NEW METHOD(S) This article describes a single incision surgical approach to the infraorbital nerve of rats and mice and the supraorbital nerve in rats for trigeminal nerve stimulation studies. This article also presents the use of cortical evoked potentials and electromyography as methods for demonstrating effective engagement between the implanted electrode and target nerve. COMPARISON WITH EXISTING METHOD(S) A number of surgical approaches to the infraorbital nerve in rats exist, many of which are technically difficult. A simple, standardized approach to infraorbital nerve in rats and mice, as well as the supraorbital nerve of rats is integral to reproducibility of future trigeminal nerve stimulation studies. CONCLUSION The infraorbital nerve of rats and mice can be easily accessed from a single dorsal incision on the bridge of the nose that avoids major anatomical structures such as the facial nerve. The supraorbital nerve is also accessible in rats from a single dorsal incision, but not mice due to size. Successful interfacing and engagement of the infra- and supraorbital nerves using the described methodology is demonstrated by recording of evoked cortical potentials and electromyography.
Collapse
Affiliation(s)
- Aaron Dingle
- Division of Plastic Surgery, Department of Surgery, University of Wisconsin-Madison, United States.
| | - Weifeng Zeng
- Division of Plastic Surgery, Department of Surgery, University of Wisconsin-Madison, United States
| | - Jared P Ness
- Department of Biomedical Engineering, University of Wisconsin-Madison, United States
| | - Nicholas Albano
- Division of Plastic Surgery, Department of Surgery, University of Wisconsin-Madison, United States
| | - Rashea L Minor
- School of Veterinary Medicine, University of Wisconsin-Madison, United States
| | - Coner Feldman
- Division of Plastic Surgery, Department of Surgery, University of Wisconsin-Madison, United States
| | - Mark Austin
- Department of Biomedical Engineering, University of Wisconsin-Madison, United States
| | - Sarah K Brodnick
- Department of Biomedical Engineering, University of Wisconsin-Madison, United States
| | - Nikita Shulzhenko
- Division of Plastic Surgery, Department of Surgery, University of Wisconsin-Madison, United States
| | - Ruston Sanchez
- Division of Plastic Surgery, Department of Surgery, University of Wisconsin-Madison, United States
| | - Wendell B Lake
- Department of Neurological Surgery, University of Wisconsin-Madison, United States
| | - Justin C Williams
- Department of Biomedical Engineering, University of Wisconsin-Madison, United States; Department of Neurological Surgery, University of Wisconsin-Madison, United States
| | - Samuel O Poore
- Division of Plastic Surgery, Department of Surgery, University of Wisconsin-Madison, United States; Department of Biomedical Engineering, University of Wisconsin-Madison, United States
| | - Aaron J Suminski
- Department of Biomedical Engineering, University of Wisconsin-Madison, United States; Department of Neurological Surgery, University of Wisconsin-Madison, United States.
| |
Collapse
|
322
|
Nurse P, Hayles J. Using genetics to understand biology. Heredity (Edinb) 2019; 123:4-13. [PMID: 31189902 PMCID: PMC6781147 DOI: 10.1038/s41437-019-0209-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
- Paul Nurse
- The Francis Crick Institute, 1, Midland Road, London, NW1 1AT, UK
| | | |
Collapse
|
323
|
Goncharov NV, Terpilowski MA, Shmurak VI, Belinskaya DA, Avdonin PV. The Rat (Rattus norvegicus) as a Model Object for Acute Organophosphate Poisoning. 1. Biochemical Aspects. J EVOL BIOCHEM PHYS+ 2019. [DOI: 10.1134/s0022093019020042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
324
|
Abstract
The underlying mechanisms that result in neurophysiological changes and cognitive sequelae in the context of repetitive mild traumatic brain injury (rmTBI) remain poorly understood. Animal models provide a unique opportunity to examine cellular and molecular responses using histological assessment, which can give important insights on the neurophysiological changes associated with the evolution of brain injury. To better understand the potential cumulative effects of multiple concussions, the focus of animal models is shifting from single to repetitive head impacts. With a growing body of literature on this subject, a review and discussion of current findings is valuable to better understand the neuropathology associated with rmTBI, to evaluate the current state of the field, and to guide future research efforts. Despite variability in experimental settings, existing animal models of rmTBI have contributed to our understanding of the underlying mechanisms following repeat concussion. However, how to reconcile the various impact methods remains one of the major challenges in the field today.
Collapse
Affiliation(s)
- Wouter S Hoogenboom
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Clinical Investigation, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA.
| | - Craig A Branch
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Physiology and Biophysics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| | - Michael L Lipton
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Departments of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| |
Collapse
|
325
|
Van Erum J, Van Dam D, De Deyn PP. PTZ-induced seizures in mice require a revised Racine scale. Epilepsy Behav 2019; 95:51-55. [PMID: 31026782 DOI: 10.1016/j.yebeh.2019.02.029] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 10/27/2022]
Abstract
Seizure severity in experimental models of epilepsy is often evaluated by means of the Racine scale, in spite of the use of seizure induction methods that are different from those of the original paper by Racine in 1972. In such cases, the use of this scale is not always justified because some seizure behaviors are significantly different from those originally described or not present at all. Correspondingly, the pentylenetetrazole (PTZ) model, which is frequently used for antiepileptic drug research, lacked an adequate assessment tool to measure seizure severity. In 2009, an adapted intensity scale for PTZ-induced seizures was already designed for rats. Here, we evaluated electroencephalographic (EEG) and behavioral parameters after a single PTZ injection, to determine whether this scale is also suitable for use in mouse studies. We found that the scale designed for rats is quite robust and can thus be applied to score seizure severity in mice. Yet, certain convulsive behaviors and EEG characteristics were distinct between species. Therefore, a species-specific scale was designed, which included the concomitant EEG characteristic next to the behavioral expressions we observed, in order to establish a user-friendly scoring scale for PTZ-induced seizures in mice. To evaluate applicability, we utilized the scale in a seizure susceptibility study of a transgenic mouse model. We demonstrated that the maximum severity scores obtained with the newly revised Racine scale highly correlated with the administered dose. Hence, the revised scale differentiates well between different classes of seizure severity.
Collapse
Affiliation(s)
- Jan Van Erum
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Antwerp, Belgium; Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Peter Paul De Deyn
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Antwerp, Belgium; Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, the Netherlands; Department of Neurology, Memory Clinic of Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.
| |
Collapse
|
326
|
Yip PK, Chapman GE, Sillito RR, Ip THR, Akhigbe G, Becker SC, Price AW, Michael-Titus AT, Armstrong JD, Tremoleda JL. Studies on long term behavioural changes in group-housed rat models of brain and spinal cord injury using an automated home cage recording system. J Neurosci Methods 2019; 321:49-63. [PMID: 30991030 DOI: 10.1016/j.jneumeth.2019.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Neurotrauma patients face major neurological sequelae. The failure in the preclinical-to-clinical translation of candidate therapies could be due to poor evaluation of rodent behaviours after neurotrauma. NEW METHOD A home cage automated system was used to study the long term behaviour of individual rats with traumatic brain injury (TBI), spinal cord injury (SCI) and non-CNS injured controls, whilst group-housed in their home cages. Naïve rats were used as baseline controls. Automated locomotor activity and body temperature recordings were carried out 24 h /day for 3 days/week during 12 weeks post-injury. Behavioural patterns, including aggression, rearing, grooming, feeding and drinking were analysed from automated video recordings during week 1, 6 and 12. RESULTS SCI animals showed a lower locomotor activity compared to TBI or control animals during light and dark phases. TBI animals showed a higher aggression during the dark phase in the first week post-injury compared to SCI or control animals. Individual grooming and rearing were reduced in SCI animals compared to TBI and control animals in the first week post-injury during the dark phase. No differences in drinking or feeding were detected between groups. Locomotor activity did not differ between naïve male and female rats, but body temperature differ between light and dark phases for both. STANDARD METHODS Injury severity was compared to standard SCI and TBI behaviour scores (BBB and mNSS, respectively) and histological analysis. CONCLUSIONS This study demonstrates the practical benefits of using a non-intrusive automated home cage recording system to observe long term individual behaviour of group-housed SCI and TBI rats.
Collapse
Affiliation(s)
- Ping K Yip
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - George E Chapman
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - T H Richard Ip
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Georgia Akhigbe
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Stephanie C Becker
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Anthony W Price
- Biological Services, Queen Mary University of London, London, United Kingdom
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - J Douglas Armstrong
- Actual Analytics Ltd, Edinburgh, United Kingdom; School of Informatics, Institute for Adaptive and Neural Computation. University of Edinburgh, Edinburgh, United Kingdom
| | - Jordi L Tremoleda
- Centre for Neuroscience, Surgery and Trauma, Centre for Trauma Sciences, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom; Biological Services, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
327
|
Bäck S, Necarsulmer J, Whitaker LR, Coke LM, Koivula P, Heathward EJ, Fortuno LV, Zhang Y, Yeh CG, Baldwin HA, Spencer MD, Mejias-Aponte CA, Pickel J, Hoffman AF, Spivak CE, Lupica CR, Underhill SM, Amara SG, Domanskyi A, Anttila JE, Airavaara M, Hope BT, Hamra FK, Richie CT, Harvey BK. Neuron-Specific Genome Modification in the Adult Rat Brain Using CRISPR-Cas9 Transgenic Rats. Neuron 2019; 102:105-119.e8. [PMID: 30792150 DOI: 10.1016/j.neuron.2019.01.035] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/13/2018] [Accepted: 01/16/2019] [Indexed: 12/28/2022]
Abstract
Historically, the rat has been the preferred animal model for behavioral studies. Limitations in genome modification have, however, caused a lag in their use compared to the bevy of available transgenic mice. Here, we have developed several transgenic tools, including viral vectors and transgenic rats, for targeted genome modification in specific adult rat neurons using CRISPR-Cas9 technology. Starting from wild-type rats, knockout of tyrosine hydroxylase was achieved with adeno-associated viral (AAV) vectors expressing Cas9 or guide RNAs (gRNAs). We subsequently created an AAV vector for Cre-dependent gRNA expression as well as three new transgenic rat lines to specifically target CRISPR-Cas9 components to dopaminergic neurons. One rat represents the first knockin rat model made by germline gene targeting in spermatogonial stem cells. The rats described herein serve as a versatile platform for making cell-specific and sequence-specific genome modifications in the adult brain and potentially other Cre-expressing tissues of the rat.
Collapse
Affiliation(s)
- Susanne Bäck
- Molecular Mechanisms of Cellular Stress and Inflammation Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Julie Necarsulmer
- Optogenetics and Transgenic Technology Core/Genetic Engineering and Viral Vector Core, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Leslie R Whitaker
- Neuronal Ensembles in Drug Addiction Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Lamarque M Coke
- Molecular Mechanisms of Cellular Stress and Inflammation Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Pyry Koivula
- Molecular Mechanisms of Cellular Stress and Inflammation Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Emily J Heathward
- Optogenetics and Transgenic Technology Core/Genetic Engineering and Viral Vector Core, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Lowella V Fortuno
- Optogenetics and Transgenic Technology Core/Genetic Engineering and Viral Vector Core, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Yajun Zhang
- Optogenetics and Transgenic Technology Core/Genetic Engineering and Viral Vector Core, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - C Grace Yeh
- Neuronal Ensembles in Drug Addiction Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Heather A Baldwin
- Molecular Mechanisms of Cellular Stress and Inflammation Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Morgan D Spencer
- Molecular Mechanisms of Cellular Stress and Inflammation Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carlos A Mejias-Aponte
- Histology Core, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - James Pickel
- Transgenic Technology Core, Intramural Research Program, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Alexander F Hoffman
- Electrophysiology Research Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Charles E Spivak
- Electrophysiology Research Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carl R Lupica
- Electrophysiology Research Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Suzanne M Underhill
- Laboratory of Molecular and Cellular Neurobiology, Intramural Research Program, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Susan G Amara
- Laboratory of Molecular and Cellular Neurobiology, Intramural Research Program, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Jenni E Anttila
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Bruce T Hope
- Neuronal Ensembles in Drug Addiction Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - F Kent Hamra
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher T Richie
- Optogenetics and Transgenic Technology Core/Genetic Engineering and Viral Vector Core, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Brandon K Harvey
- Molecular Mechanisms of Cellular Stress and Inflammation Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA; Optogenetics and Transgenic Technology Core/Genetic Engineering and Viral Vector Core, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
328
|
Coffey KR, Marx RE, Neumaier JF. DeepSqueak: a deep learning-based system for detection and analysis of ultrasonic vocalizations. Neuropsychopharmacology 2019; 44:859-868. [PMID: 30610191 PMCID: PMC6461910 DOI: 10.1038/s41386-018-0303-6] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/04/2018] [Accepted: 12/16/2018] [Indexed: 01/11/2023]
Abstract
Rodents engage in social communication through a rich repertoire of ultrasonic vocalizations (USVs). Recording and analysis of USVs has broad utility during diverse behavioral tests and can be performed noninvasively in almost any rodent behavioral model to provide rich insights into the emotional state and motor function of the test animal. Despite strong evidence that USVs serve an array of communicative functions, technical and financial limitations have been barriers for most laboratories to adopt vocalization analysis. Recently, deep learning has revolutionized the field of machine hearing and vision, by allowing computers to perform human-like activities including seeing, listening, and speaking. Such systems are constructed from biomimetic, "deep", artificial neural networks. Here, we present DeepSqueak, a USV detection and analysis software suite that can perform human quality USV detection and classification automatically, rapidly, and reliably using cutting-edge regional convolutional neural network architecture (Faster-RCNN). DeepSqueak was engineered to allow non-experts easy entry into USV detection and analysis yet is flexible and adaptable with a graphical user interface and offers access to numerous input and analysis features. Compared to other modern programs and manual analysis, DeepSqueak was able to reduce false positives, increase detection recall, dramatically reduce analysis time, optimize automatic syllable classification, and perform automatic syntax analysis on arbitrarily large numbers of syllables, all while maintaining manual selection review and supervised classification. DeepSqueak allows USV recording and analysis to be added easily to existing rodent behavioral procedures, hopefully revealing a wide range of innate responses to provide another dimension of insights into behavior when combined with conventional outcome measures.
Collapse
Affiliation(s)
- Kevin R. Coffey
- 0000000122986657grid.34477.33Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA 98104 USA
| | - Ruby E. Marx
- 0000000122986657grid.34477.33Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA 98104 USA
| | - John F. Neumaier
- 0000000122986657grid.34477.33Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA 98104 USA
| |
Collapse
|
329
|
Pelch KE, Bolden AL, Kwiatkowski CF. Environmental Chemicals and Autism: A Scoping Review of the Human and Animal Research. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:46001. [PMID: 30942615 PMCID: PMC6785231 DOI: 10.1289/ehp4386] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 05/23/2023]
Abstract
BACKGROUND Estimates of autism prevalence have increased dramatically over the past two decades. Evidence suggests environmental factors may contribute to the etiology of the disorder. OBJECTIVES This scoping review aimed to identify and categorize primary research and reviews on the association between prenatal and early postnatal exposure to environmental chemicals and the development of autism in epidemiological studies and rodent models of autism. METHODS PubMed was searched through 8 February 2018. Included studies assessed exposure to environmental chemicals prior to 2 months of age in humans or 14 d in rodents. Rodent studies were considered relevant if they included at least one measurement of reciprocal social communicative behavior or repetitive and stereotyped behavior. Study details are presented in interactive displays using Tableau Public. RESULTS The search returned 21,603 unique studies, of which 54 epidemiological studies, 46 experimental rodent studies, and 50 reviews were deemed relevant, covering 152 chemical exposures. The most frequently studied exposures in humans were particulate matter ([Formula: see text]), mercury ([Formula: see text]), nonspecific air pollution ([Formula: see text]), and lead ([Formula: see text]). In rodent studies, the most frequently studied exposures were chlorpyrifos ([Formula: see text]), mercury ([Formula: see text]), and lead ([Formula: see text]). DISCUSSION Although research is growing rapidly, wide variability exists in study design and conduct, exposures investigated, and outcomes assessed. Conclusions focus on recommendations to guide development of best practices in epidemiology and toxicology, including greater harmonization across these fields of research to more quickly and efficiently identify chemicals of concern. In particular, we recommend chlorpyrifos, lead, and polychlorinated biphenyls (PCBs) be systematically reviewed in order to assess their relationship with the development of autism. There is a pressing need to move forward quickly and efficiently to understand environmental influences on autism in order to answer current regulatory questions and inform treatment and prevention efforts. https://doi.org/10.1289/EHP4386.
Collapse
Affiliation(s)
| | | | - Carol F. Kwiatkowski
- The Endocrine Disruption Exchange, Eckert, Colorado, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
330
|
Electrophysiological Properties of Medium Spiny Neuron Subtypes in the Caudate-Putamen of Prepubertal Male and Female Drd1a-tdTomato Line 6 BAC Transgenic Mice. eNeuro 2019; 6:eN-CFN-0016-19. [PMID: 30899778 PMCID: PMC6426437 DOI: 10.1523/eneuro.0016-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/12/2019] [Accepted: 02/24/2019] [Indexed: 12/21/2022] Open
Abstract
The caudate-putamen is a striatal brain region essential for sensorimotor behaviors, habit learning, and other cognitive and premotor functions. The output and predominant neuron of the caudate-putamen is the medium spiny neuron (MSN). MSNs present discrete cellular subtypes that show differences in neurochemistry, dopamine receptor expression, efferent targets, gene expression, functional roles, and most importantly for this study, electrophysiological properties. MSN subtypes include the striatonigral and the striatopallidal groups. Most studies identify the striatopallidal MSN subtype as being more excitable than the striatonigral MSN subtype. However, there is some divergence between studies regarding the exact differences in electrophysiological properties. Furthermore, MSN subtype electrophysiological properties have not been reported disaggregated by biological sex. We addressed these questions using prepubertal male and female Drd1a-tdTomato line 6 BAC transgenic mice, an important transgenic line that has not yet received extensive electrophysiological analysis. We made acute caudate-putamen brain slices and assessed a robust battery of 16 relevant electrophysiological properties using whole-cell patch-clamp recording, including intrinsic membrane, action potential, and miniature EPSC (mEPSC) properties. We found that: (1) MSN subtypes exhibited multiple differential electrophysiological properties in both sexes, including rheobase, action potential threshold and width, input resistance in both the linear and rectified ranges, and mEPSC amplitude; (2) select electrophysiological properties showed interactions between MSN subtype and sex. These findings provide a comprehensive evaluation of mouse caudate-putamen MSN subtype electrophysiological properties across females and males, both confirming and extending previous studies.
Collapse
|
331
|
Cuello AC, Hall H, Do Carmo S. Experimental Pharmacology in Transgenic Rodent Models of Alzheimer's Disease. Front Pharmacol 2019; 10:189. [PMID: 30886583 PMCID: PMC6409318 DOI: 10.3389/fphar.2019.00189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/14/2019] [Indexed: 12/15/2022] Open
Abstract
This Mini Review discusses the merits and shortfalls of transgenic (tg) rodents modeling aspects of the human Alzheimer’s disease (AD) pathology and their application to evaluate experimental therapeutics. It addresses some of the differences between mouse and rat tg models for these investigations. It relates, in a condensed fashion, the experience of our research laboratory with the application of anti-inflammatory compounds and S-adenosylmethionine (SAM) at the earliest stages of AD-like amyloid pathology in tg mice. The application of SAM was intended to revert the global brain DNA hypomethylation unleashed by the intraneuronal accumulation of amyloid-β-immunoreactive material, an intervention that restored levels of DNA methylation including of the bace1 gene. This review also summarizes experimental pharmacology observations made in the McGill tg rat model of AD-like pathology by applying “nano-lithium” or a drug with allosteric M1 muscarinic and sigma 1 receptor agonistic properties (AF710B). Extremely low doses of lithium (up to 400 times lower than used in the clinic) had remarkable beneficial effects on lowering pathology and improving cognitive functions in tg rats. Likewise, AF710B treatment, even at advanced stages of the pathology, displayed remarkable beneficial effects. This drug, in experimental conditions, demonstrated possible “disease-modifying” properties as pathology was frankly diminished and cognition improved after a month of “wash-out” period. The Mini-Review ends with a discussion on the predictive value of similar experimental pharmacological interventions in current rodent tg models. It comments on the validity of some of these approaches for early interventions at preclinical stages of AD, interventions which may be envisioned once definitive diagnosis of AD before clinical presentation is made possible.
Collapse
Affiliation(s)
- A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Hélène Hall
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| |
Collapse
|
332
|
Jackson TC, Kochanek PM. A New Vision for Therapeutic Hypothermia in the Era of Targeted Temperature Management: A Speculative Synthesis. Ther Hypothermia Temp Manag 2019; 9:13-47. [PMID: 30802174 PMCID: PMC6434603 DOI: 10.1089/ther.2019.0001] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Three decades of animal studies have reproducibly shown that hypothermia is profoundly cerebroprotective during or after a central nervous system (CNS) insult. The success of hypothermia in preclinical acute brain injury has not only fostered continued interest in research on the classic secondary injury mechanisms that are prevented or blunted by hypothermia but has also sparked a surge of new interest in elucidating beneficial signaling molecules that are increased by cooling. Ironically, while research into cold-induced neuroprotection is enjoying newfound interest in chronic neurodegenerative disease, conversely, the scope of the utility of therapeutic hypothermia (TH) across the field of acute brain injury is somewhat controversial and remains to be fully defined. This has led to the era of Targeted Temperature Management, which emphasizes a wider range of temperatures (33–36°C) showing benefit in acute brain injury. In this comprehensive review, we focus on our current understandings of the novel neuroprotective mechanisms activated by TH, and discuss the critical importance of developmental age germane to its clinical efficacy. We review emerging data on four cold stress hormones and three cold shock proteins that have generated new interest in hypothermia in the field of CNS injury, to create a framework for new frontiers in TH research. We make the case that further elucidation of novel cold responsive pathways might lead to major breakthroughs in the treatment of acute brain injury, chronic neurological diseases, and have broad potential implications for medicines of the distant future, including scenarios such as the prevention of adverse effects of long-duration spaceflight, among others. Finally, we introduce several new phrases that readily summarize the essence of the major concepts outlined by this review—namely, Ultramild Hypothermia, the “Responsivity of Cold Stress Pathways,” and “Hypothermia in a Syringe.”
Collapse
Affiliation(s)
- Travis C Jackson
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
333
|
The Application of Adeno-Associated Viral Vector Gene Therapy to the Treatment of Fragile X Syndrome. Brain Sci 2019; 9:brainsci9020032. [PMID: 30717399 PMCID: PMC6406794 DOI: 10.3390/brainsci9020032] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 01/18/2023] Open
Abstract
Viral vector-mediated gene therapy has grown by leaps and bounds over the past several years. Although the reasons for this progress are varied, a deeper understanding of the basic biology of the viruses, the identification of new and improved versions of viral vectors, and simply the vast experience gained by extensive testing in both animal models of disease and in clinical trials, have been key factors. Several studies have investigated the efficacy of adeno-associated viral (AAV) vectors in the mouse model of fragile X syndrome where AAVs have been used to express fragile X mental retardation protein (FMRP), which is missing or highly reduced in the disorder. These studies have demonstrated a range of efficacies in different tests from full correction, to partial rescue, to no effect. Here we provide a backdrop of recent advances in AAV gene therapy as applied to central nervous system disorders, outline the salient features of the fragile X studies, and discuss several key issues for moving forward. Collectively, the findings to date from the mouse studies on fragile X syndrome, and data from clinical trials testing AAVs in other neurological conditions, indicate that AAV-mediated gene therapy could be a viable strategy for treating fragile X syndrome.
Collapse
|
334
|
Shi SQ, Johnson CH. Circadian biology and sleep in monogenic neurological disorders and its potential application in drug discovery. Curr Opin Behav Sci 2019; 25:23-30. [PMID: 31289731 PMCID: PMC6615557 DOI: 10.1016/j.cobeha.2018.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Sleep disturbances are common in people with monogenic neurological disorders and they dramatically affect the life of individuals with the disorders and their families. The associated sleep problems are probably caused by multiple factors that have not been elucidated. Study of the underlying molecular cause, behavioral phenotypes, and reciprocal interactions in several single-gene disorders (Angelman Syndrome, Fragile X Syndrome, Rett Syndrome, and Huntington's Disease) leads to the suggestion that sleep disruption and other symptoms may directly result from abnormal operation of circadian systems due to genetic alteration and/or conflicting environmental cues for clock entrainment. Therefore, because circadian patterns modify the symptoms of neurological disorders, treatments that modulate our daily rhythms may identify heretofore unappreciated therapies for the underlying disorders.
Collapse
Affiliation(s)
- Shu-Qun Shi
- Department of Biological Sciences, Vanderbilt University, USA
| | - Carl Hirschie Johnson
- Department of Biological Sciences, Vanderbilt University, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical School, USA
| |
Collapse
|
335
|
Hake HS, Davis JKP, Wood RR, Tanner MK, Loetz EC, Sanchez A, Ostrovskyy M, Oleson EB, Grigsby J, Doblin R, Greenwood BN. 3,4-methylenedioxymethamphetamine (MDMA) impairs the extinction and reconsolidation of fear memory in rats. Physiol Behav 2019; 199:343-350. [PMID: 30529341 PMCID: PMC6557441 DOI: 10.1016/j.physbeh.2018.12.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 11/30/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023]
Abstract
Clinical trials have demonstrated that 3,4-methylenedioxymethamphetamine (MDMA) paired with psychotherapy is more effective at reducing symptoms of post-traumatic stress disorder (PTSD) than psychotherapy or pharmacotherapy, alone or in combination. The processes through which MDMA acts to enhance psychotherapy are not well understood. Given that fear memories contribute to PTSD symptomology, MDMA could augment psychotherapy by targeting fear memories. The current studies investigated the effects of a single administration of MDMA on extinction and reconsolidation of cued and contextual fear memory in adult, male Long-Evans rats. Rats were exposed to contextual or auditory fear conditioning followed by systemic administration of saline or varying doses of MDMA (between 1 and 10 mg/kg) either 30 min before fear extinction training or immediately after brief fear memory retrieval (i.e. during the reconsolidation phase). MDMA administered prior to fear extinction training failed to enhance fear extinction memory, and in fact impaired drug-free cued fear extinction recall without impacting later fear relapse. MDMA administered during the reconsolidation phase, but not outside of the reconsolidation phase, produced a delayed and persistent reduction in conditioned fear. These findings are consistent with a general memory-disrupting effect of MDMA and suggest that MDMA could augment psychotherapy by modifying fear memories during reconsolidation without necessarily enhancing their extinction.
Collapse
Affiliation(s)
- Holly S Hake
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - Jazmyne K P Davis
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - River R Wood
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - Margaret K Tanner
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - Esteban C Loetz
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - Anais Sanchez
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - Mykola Ostrovskyy
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - Erik B Oleson
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| | - Jim Grigsby
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA; Department of Medicine, University of Colorado Denver School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Rick Doblin
- Multidisciplinary Association for Psychedelic Studies, 1115 Mission Street, Santa Cruz, CA 95060-9989, USA
| | - Benjamin N Greenwood
- Department of Psychology, University of Colorado Denver, PO Box 173364, Denver, CO 80217-3364, USA.
| |
Collapse
|
336
|
Kisko TM, Braun MD, Michels S, Witt SH, Rietschel M, Culmsee C, Schwarting RKW, Wöhr M. Sex‐dependent effects of
Cacna1c
haploinsufficiency on juvenile social play behavior and pro‐social 50‐kHz ultrasonic communication in rats. GENES BRAIN AND BEHAVIOR 2019; 19:e12552. [DOI: 10.1111/gbb.12552] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Theresa M. Kisko
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of PsychologyPhilipps‐Universität Marburg Marburg Germany
| | - Moria D. Braun
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of PsychologyPhilipps‐Universität Marburg Marburg Germany
| | - Susanne Michels
- Institute of Pharmacology and Clinical PharmacyPhilipps‐Universität Marburg Marburg Germany
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Faculty of Medicine MannheimRuprecht‐Karls‐Universität Heidelberg Mannheim Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Faculty of Medicine MannheimRuprecht‐Karls‐Universität Heidelberg Mannheim Germany
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical PharmacyPhilipps‐Universität Marburg Marburg Germany
- Center for Mind, Brain, and Behavior (CMBB)Philipps‐Universität Marburg Marburg Germany
| | - Rainer K. W. Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of PsychologyPhilipps‐Universität Marburg Marburg Germany
- Center for Mind, Brain, and Behavior (CMBB)Philipps‐Universität Marburg Marburg Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of PsychologyPhilipps‐Universität Marburg Marburg Germany
- Center for Mind, Brain, and Behavior (CMBB)Philipps‐Universität Marburg Marburg Germany
| |
Collapse
|
337
|
Genetic labeling reveals temporal and spatial expression pattern of D2 dopamine receptor in rat forebrain. Brain Struct Funct 2019; 224:1035-1049. [PMID: 30604007 PMCID: PMC6499762 DOI: 10.1007/s00429-018-01824-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/20/2018] [Indexed: 01/11/2023]
Abstract
The D2 dopamine receptor (Drd2) is implicated in several brain disorders such as schizophrenia, Parkinson’s disease, and drug addiction. Drd2 is also the primary target of both antipsychotics and Parkinson’s disease medications. Although the expression pattern of Drd2 is relatively well known in mouse brain, the temporal and spatial distribution of Drd2 is lesser clear in rat brain due to the lack of Drd2 reporter rat lines. Here, we used CRISPR/Cas9 techniques to generate two knockin rat lines: Drd2::Cre and Rosa26::loxp-stop-loxp-tdTomato. By crossing these two lines, we produced Drd2 reporter rats expressing the fluorescence protein tdTomato under the control of the endogenous Drd2 promoter. Using fluorescence imaging and unbiased stereology, we revealed the cellular expression pattern of Drd2 in adult and postnatal rat forebrain. Strikingly, the Drd2 expression pattern differs between Drd2 reporter rats and Drd2 reporter mice generated by BAC transgene in prefrontal cortex and hippocampus. These results provide fundamental information needed for the study of Drd2 function in rat forebrain. The Drd2::Cre rats generated here may represent a useful tool to study the function of neuronal populations expressing Drd2.
Collapse
|
338
|
Savell KE, Bach SV, Zipperly ME, Revanna JS, Goska NA, Tuscher JJ, Duke CG, Sultan FA, Burke JN, Williams D, Ianov L, Day JJ. A Neuron-Optimized CRISPR/dCas9 Activation System for Robust and Specific Gene Regulation. eNeuro 2019; 6:ENEURO.0495-18.2019. [PMID: 30863790 PMCID: PMC6412672 DOI: 10.1523/eneuro.0495-18.2019] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 12/19/2022] Open
Abstract
CRISPR-based technology has provided new avenues to interrogate gene function, but difficulties in transgene expression in post-mitotic neurons has delayed incorporation of these tools in the central nervous system (CNS). Here, we demonstrate a highly efficient, neuron-optimized dual lentiviral CRISPR-based transcriptional activation (CRISPRa) system capable of robust, modular, and tunable gene induction and multiplexed gene regulation across several primary rodent neuron culture systems. CRISPRa targeting unique promoters in the complex multi-transcript gene brain-derived neurotrophic factor (Bdnf) revealed both transcript- and genome-level selectivity of this approach, in addition to highlighting downstream transcriptional and physiological consequences of Bdnf regulation. Finally, we illustrate that CRISPRa is highly efficient in vivo, resulting in increased protein levels of a target gene in diverse brain structures. Taken together, these results demonstrate that CRISPRa is an efficient and selective method to study gene expression programs in brain health and disease.
Collapse
Affiliation(s)
- Katherine E. Savell
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Svitlana V. Bach
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Morgan E. Zipperly
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Jasmin S. Revanna
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Nicholas A. Goska
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Jennifer J. Tuscher
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Corey G. Duke
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Faraz A. Sultan
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Julia N. Burke
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Derek Williams
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Jeremy J. Day
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, 35294
| |
Collapse
|
339
|
Kelly SC, McKay EC, Beck JS, Collier TJ, Dorrance AM, Counts SE. Locus Coeruleus Degeneration Induces Forebrain Vascular Pathology in a Transgenic Rat Model of Alzheimer's Disease. J Alzheimers Dis 2019; 70:371-388. [PMID: 31177220 PMCID: PMC6929678 DOI: 10.3233/jad-190090] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Noradrenergic locus coeruleus (LC) neuron loss is a significant feature of mild cognitive impairment and Alzheimer's disease (AD). The LC is the primary source of norepinephrine in the forebrain, where it modulates attention and memory in vulnerable cognitive regions such as prefrontal cortex (PFC) and hippocampus. Furthermore, LC-mediated norepinephrine signaling is thought to play a role in blood-brain barrier (BBB) maintenance and neurovascular coupling, suggesting that LC degeneration may impact the high comorbidity of cerebrovascular disease and AD. However, the extent to which LC projection system degeneration influences vascular pathology is not fully understood. To address this question in vivo, we stereotactically lesioned LC projection neurons innervating the PFC of six-month-old Tg344-19 AD rats using the noradrenergic immunotoxin, dopamine-β-hydroxylase IgG-saporin (DBH-sap), or an untargeted control IgG-saporin (IgG-sap). DBH-sap-lesioned animals performed significantly worse than IgG-sap animals on the Barnes maze task in measures of both spatial and working memory. DBH-sap-lesioned rats also displayed increased amyloid and inflammation pathology compared to IgG-sap controls. However, we also discovered prominent parenchymal albumin extravasation with DBH-sap lesions indicative of BBB breakdown. Moreover, microvessel wall-to-lumen ratios were increased in the PFC of DBH-sap compared to IgG-sap rats, suggesting that LC deafferentation results in vascular remodeling. Finally, we noted an early emergence of amyloid angiopathy in the DBH-sap-lesioned Tg344-19 AD rats. Taken together, these data indicate that LC projection system degeneration is a nexus lesion that compromises both vascular and neuronal function in cognitive brain areas during the prodromal stages of AD.
Collapse
Affiliation(s)
- Sarah C. Kelly
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
| | - Erin C. McKay
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - John S. Beck
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Timothy J. Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Anne M. Dorrance
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Scott E. Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, MI, USA
- Hauenstein Neurosciences Center, Mercy Health Saint Mary’s Hospital, Grand Rapids, MI, USA
- Michigan Alzheimer’s Disease Core Center, Ann Arbor, MI, USA
| |
Collapse
|
340
|
Beery AK. Frank Beach award winner: Neuroendocrinology of group living. Horm Behav 2019; 107:67-75. [PMID: 30439353 PMCID: PMC6371784 DOI: 10.1016/j.yhbeh.2018.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/31/2018] [Accepted: 11/09/2018] [Indexed: 01/02/2023]
Abstract
Why do members of some species live in groups while others are solitary? Group living (sociality) has often been studied from an evolutionary perspective, but less is known about the neurobiology of affiliation outside the realms of mating and parenting. Colonial species offer a valuable opportunity to study nonsexual affiliative behavior between adult peers. Meadow voles (Microtus pennsylvanicus) display environmentally induced variation in social behavior, maintaining exclusive territories in summer months, but living in social groups in winter. Research on peer relationships in female meadow voles demonstrates that these selective preferences are mediated differently than mate relationships in socially monogamous prairie voles, but are also impacted by oxytocin and HPA axis signaling. This review addresses day-length dependent variation in physiology and behavior, and presents the current understanding of the mechanisms supporting selective social relationships in meadow voles, with connections to lessons from other species.
Collapse
Affiliation(s)
- Annaliese K Beery
- Department of Psychology, Department of Biology, Program in Neuroscience, Smith College, Northampton, MA 01063, United States of America.
| |
Collapse
|
341
|
Experimental Models of Tauopathy - From Mechanisms to Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:381-391. [PMID: 32096051 DOI: 10.1007/978-981-32-9358-8_28] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Animal models have been instrumental in reproducing key aspects of human tauopathy. In pursuing these efforts, the mouse continues to have a prominent role. In this chapter, we focus on models that overexpress wild-type or mutant forms of tau, the latter being based on mutations found in familial cases of frontotemporal dementia. We review some of these models in more detail and discuss what they have revealed about the underlying pathomechanisms, as well as highlighting new developments that exploit gene editing tools such as TALEN and CRISPR. Interestingly, when investigating the role of tau in impairing cellular functions, common themes emerge. Because tau is a scaffolding protein that aggregates in the somatodendritic domain under pathological conditions, it traps proteins such as parkin and JIP1, preventing them from executing their normal function in mitophagy and axonal transport, respectively. Another aspect is the emerging role of tau in the translational machinery and the finding that the somatodendritic accumulation of tau in Alzheimer's disease may in part be due to the induction of the de novo synthesis of tau by amyloid-β via the Fyn/ERK/S6 pathway. We further discuss treatment strategies such as tau-based vaccinations and therapeutic ultrasound and conclude by discussing whether there is a future for animal models of tauopathies.
Collapse
|
342
|
Breger LS, Fuzzati Armentero MT. Genetically engineered animal models of Parkinson's disease: From worm to rodent. Eur J Neurosci 2018; 49:533-560. [PMID: 30552719 DOI: 10.1111/ejn.14300] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is a progressive neurological disorder characterised by aberrant accumulation of insoluble proteins, including alpha-synuclein, and a loss of dopaminergic neurons in the substantia nigra. The extended neurodegeneration leads to a drop of striatal dopamine levels responsible for disabling motor and non-motor impairments. Although the causes of the disease remain unclear, it is well accepted among the scientific community that the disorder may also have a genetic component. For that reason, the number of genetically engineered animal models has greatly increased over the past two decades, ranging from invertebrates to more complex organisms such as mice and rats. This trend is growing as new genetic variants associated with the disease are discovered. The EU Joint Programme - Neurodegenerative Disease Research (JPND) has promoted the creation of an online database aiming at summarising the different features of experimental models of Parkinson's disease. This review discusses available genetic models of PD and the extent to which they adequately mirror the human pathology and reflects on future development and uses of genetically engineered experimental models for the study of PD.
Collapse
Affiliation(s)
- Ludivine S Breger
- Institut des Maladies Neurodégénératives, CNRS UMR 5293, Centre Broca Nouvelle Aquitaine, Université de Bordeaux, Bordeaux cedex, France
| | | |
Collapse
|
343
|
Species-dependent extracranial manifestations of a brain seeking breast cancer cell line. PLoS One 2018; 13:e0208340. [PMID: 30532191 PMCID: PMC6287854 DOI: 10.1371/journal.pone.0208340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/15/2018] [Indexed: 11/19/2022] Open
Abstract
PURPOSE Metastatic brain tumors pose a severe problem in the treatment of patients with breast carcinoma. Preclinical models have been shown to play an important role in unraveling the underlying mechanisms behind the metastatic process and evaluation of new therapeutic approaches. As the size of the rat brain allows improved in vivo imaging, we attempted to establish a rat model for breast cancer brain metastasis that allows follow-up by 7 tesla (7T) preclinical Magnetic Resonance Imaging (MRI). PROCEDURES Green fluorescent protein-transduced (eGFP) MDA-MB-231br breast cancer cells were labeled with micron-sized particles of iron oxide (MPIOs) and intracardially injected in the left ventricle of female nude rats and mice. 7T preclinical MRI was performed to show the initial distribution of MPIO-labeled cancer cells and to visualize metastasis in the brain. Occurrence of potential metastasis outside the brain was evaluated by 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET)/computed tomography (CT) and potential bone lesions were assessed using [18F]sodium fluoride ([18F]NaF) PET/CT. RESULTS The first signs of brain metastasis development were visible as hyperintensities on T2-weighted (T2w) MR images acquired 3 weeks after intracardiac injection in rats and mice. Early formation of unexpected bone metastasis in rats was clinically observed and assessed using PET/CT. Almost no bone metastasis development was observed in mice after PET/CT evaluation. CONCLUSIONS Our results suggest that the metastatic propensity of the MDA-MB-231br/eGFP cancer cell line outside the brain is species-dependent. Because of early and abundant formation of bone metastasis with the MDA-MB-231br/eGFP cancer cell line, this rat model is currently not suitable for investigating brain metastasis as a single disease model nor for evaluation of novel brain metastasis treatment strategies.
Collapse
|
344
|
Sieveritz B, García-Muñoz M, Arbuthnott GW. Thalamic afferents to prefrontal cortices from ventral motor nuclei in decision-making. Eur J Neurosci 2018; 49:646-657. [PMID: 30346073 PMCID: PMC6587977 DOI: 10.1111/ejn.14215] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 07/19/2018] [Accepted: 07/24/2018] [Indexed: 01/23/2023]
Abstract
The focus of this literature review is on the three interacting brain areas that participate in decision‐making: basal ganglia, ventral motor thalamic nuclei, and medial prefrontal cortex, with an emphasis on the participation of the ventromedial and ventral anterior motor thalamic nuclei in prefrontal cortical function. Apart from a defining input from the mediodorsal thalamus, the prefrontal cortex receives inputs from ventral motor thalamic nuclei that combine to mediate typical prefrontal functions such as associative learning, action selection, and decision‐making. Motor, somatosensory and medial prefrontal cortices are mainly contacted in layer 1 by the ventral motor thalamic nuclei and in layer 3 by thalamocortical input from mediodorsal thalamus. We will review anatomical, electrophysiological, and behavioral evidence for the proposed participation of ventral motor thalamic nuclei and medial prefrontal cortex in rat and mouse motor decision‐making.
Collapse
Affiliation(s)
- Bianca Sieveritz
- Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| | - Marianela García-Muñoz
- Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| | - Gordon W Arbuthnott
- Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| |
Collapse
|
345
|
Davis J, Xu F, Hatfield J, Lee H, Hoos MD, Popescu D, Crooks E, Kim R, Smith SO, Robinson JK, Benveniste H, Van Nostrand WE. A Novel Transgenic Rat Model of Robust Cerebral Microvascular Amyloid with Prominent Vasculopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2877-2889. [PMID: 30446159 PMCID: PMC6334267 DOI: 10.1016/j.ajpath.2018.07.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 10/27/2022]
Abstract
Accumulation of fibrillar amyloid β protein in blood vessels of the brain, a condition known as cerebral amyloid angiopathy (CAA), is a common pathology of elderly individuals, a prominent comorbidity of Alzheimer disease, and a driver of vascular cognitive impairment and dementia. Although several transgenic mouse strains have been generated that develop varying levels of CAA, consistent models of associated cerebral microhemorrhage and vasculopathy observed clinically have been lacking. Reliable preclinical animal models of CAA and microhemorrhage are needed to investigate the molecular pathogenesis of this condition. Herein, we describe the generation and characterization of a novel transgenic rat (rTg-DI) that produces low levels of human familial CAA Dutch/Iowa E22Q/D23N mutant amyloid β protein in brain and faithfully recapitulates many of the pathologic aspects of human small-vessel CAA. rTg-DI rats exhibit early-onset and progressive accumulation of cerebral microvascular fibrillar amyloid accompanied by early-onset and sustained behavioral deficits. Comparable to CAA in humans, the cerebral microvascular amyloid in rTg-DI rats causes capillary structural alterations, promotes prominent perivascular neuroinflammation, and produces consistent, robust microhemorrhages and small-vessel occlusions that are readily detected by magnetic resonance imaging. The rTg-DI rats provide a new model to investigate the pathogenesis of small-vessel CAA and microhemorrhages, to develop effective biomarkers for this condition and to test therapeutic interventions.
Collapse
Affiliation(s)
- Judianne Davis
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
| | - Feng Xu
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
| | - Joshua Hatfield
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
| | - Hedok Lee
- Department of Anesthesiology, Yale University, New Haven, Connecticut
| | - Michael D Hoos
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York
| | - Dominique Popescu
- Department of Psychology, Stony Brook University, Stony Brook, New York
| | - Elliot Crooks
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York
| | - Regina Kim
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
| | - Steven O Smith
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York
| | - John K Robinson
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Psychology, University of Rhode Island, Kingston, Rhode Island
| | - Helene Benveniste
- Department of Anesthesiology, Yale University, New Haven, Connecticut
| | - William E Van Nostrand
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island.
| |
Collapse
|
346
|
Drozd HP, Karathanasis SF, Molosh AI, Lukkes JL, Clapp DW, Shekhar A. From bedside to bench and back: Translating ASD models. PROGRESS IN BRAIN RESEARCH 2018; 241:113-158. [PMID: 30447753 DOI: 10.1016/bs.pbr.2018.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism spectrum disorders (ASD) represent a heterogeneous group of disorders defined by deficits in social interaction/communication and restricted interests, behaviors, or activities. Models of ASD, developed based on clinical data and observations, are used in basic science, the "bench," to better understand the pathophysiology of ASD and provide therapeutic options for patients in the clinic, the "bedside." Translational medicine creates a bridge between the bench and bedside that allows for clinical and basic science discoveries to challenge one another to improve the opportunities to bring novel therapies to patients. From the clinical side, biomarker work is expanding our understanding of possible mechanisms of ASD through measures of behavior, genetics, imaging modalities, and serum markers. These biomarkers could help to subclassify patients with ASD in order to better target treatments to a more homogeneous groups of patients most likely to respond to a candidate therapy. In turn, basic science has been responding to developments in clinical evaluation by improving bench models to mechanistically and phenotypically recapitulate the ASD phenotypes observed in clinic. While genetic models are identifying novel therapeutics targets at the bench, the clinical efforts are making progress by defining better outcome measures that are most representative of meaningful patient responses. In this review, we discuss some of these challenges in translational research in ASD and strategies for the bench and bedside to bridge the gap to achieve better benefits to patients.
Collapse
Affiliation(s)
- Hayley P Drozd
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sotirios F Karathanasis
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrei I Molosh
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jodi L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - D Wade Clapp
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anantha Shekhar
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
347
|
Mecheri B, Paris F, Lübbert H. Histological investigations on the dura mater vascular system of mice. Acta Histochem 2018; 120:846-857. [PMID: 30292321 DOI: 10.1016/j.acthis.2018.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/06/2018] [Accepted: 09/27/2018] [Indexed: 11/25/2022]
Abstract
The human dura mater encephali is a well innervated and vascularized membrane. Its vascular system plays a crucial role in disorders and pathological cases like dural hematoma, meningitis, and different headache types. To investigate these diseases mouse models are increasingly being used. However, the literature on the vascular system of the mouse dura mater is sparse and explicit studies concerned exclusively with its vasculature are lacking. Here we present a detailed light and scanning electron microscopic investigation of the supratentorial dura mater of the mouse species, with a focus on the largest part of it, the parietal dura mater. By utilizing different immunohistochemical and classical staining methods, a "cartography" of the vascular system was achieved. Additionally, the different blood vessel types with their mural cells were characterized. In contrast to humans, no arteries were found in the mouse parietal dura mater. Its supply is assured through frontolateral and occipital localized arteriolar branches. These arteriolar vessels exhibit in some specimens arteriolar anastomoses with one another. The venous blood is drained to the superior sagittal and transverse sinus through satellite venules accompanying the arterioles or through solitary venules. In all samples, large ruptured venules were identified in the frontolateral dural area. Scanning electron microscopy revealed that these vessels were ruptured on the dorsal side (skull bones-oriented side) of the dura. Our results contribute to the anatomical data on the mouse species and may set up a basis for fundamental investigation of disorders, for which the role of dural blood vessels is not yet clarified.
Collapse
|
348
|
Bruckner DM, Connerney JJ, Dordick JS. Advancing in vitro
- in vivo
toxicity correlations via high-throughput three-dimensional primary hepatocyte culture. AIChE J 2018. [DOI: 10.1002/aic.16442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Dylan M. Bruckner
- Dept. of Chemical and Biological Engineering, Center for Biotechnology & Interdisciplinary Studies; Rensselaer Polytechnic Institute; Troy NY, 12180
| | | | - Jonathan S. Dordick
- Dept. of Chemical and Biological Engineering, Center for Biotechnology & Interdisciplinary Studies; Rensselaer Polytechnic Institute; Troy NY, 12180
| |
Collapse
|
349
|
Gargiulo G. Next-Generation in vivo Modeling of Human Cancers. Front Oncol 2018; 8:429. [PMID: 30364119 PMCID: PMC6192385 DOI: 10.3389/fonc.2018.00429] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022] Open
Abstract
Animal models of human cancers played a major role in our current understanding of tumor biology. In pre-clinical oncology, animal models empowered drug target and biomarker discovery and validation. In turn, this resulted in improved care for cancer patients. In the quest for understanding and treating a diverse spectrum of cancer types, technological breakthroughs in genetic engineering and single cell "omics" offer tremendous potential to enhance the informative value of pre-clinical models. Here, I review the state-of-the-art in modeling human cancers with focus on animal models for human malignant gliomas. The review highlights the use of glioma models in dissecting mechanisms of tumor initiation, in the retrospective identification of tumor cell-of-origin, in understanding tumor heterogeneity and in testing the potential of immuno-oncology. I build on the deep review of glioma models as a basis for a more general discussion of the potential ways in which transformative technologies may shape the next-generation of pre-clinical models. I argue that refining animal models along the proposed lines will benefit the success rate of translation for pre-clinical research in oncology.
Collapse
Affiliation(s)
- Gaetano Gargiulo
- Molecular Oncology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
350
|
Scheggi S, De Montis MG, Gambarana C. Making Sense of Rodent Models of Anhedonia. Int J Neuropsychopharmacol 2018; 21:1049-1065. [PMID: 30239762 PMCID: PMC6209858 DOI: 10.1093/ijnp/pyy083] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 09/18/2018] [Indexed: 01/04/2023] Open
Abstract
A markedly reduced interest or pleasure in activities previously considered pleasurable is a main symptom in mood disorder and psychosis and is often present in other psychiatric disorders and neurodegenerative diseases. This condition can be labeled as "anhedonia," although in its most rigorous connotation the term refers to the lost capacity to feel pleasure that is one aspect of the complex phenomenon of processing and responding to reward. The responses to rewarding stimuli are relatively easy to study in rodents, and the experimental conditions that consistently and persistently impair these responses are used to model anhedonia. To this end, long-term exposure to environmental aversive conditions is primarily used, and the resulting deficits in reward responses are often accompanied by other deficits that are mainly reminiscent of clinical depressive symptoms. The different components of impaired reward responses induced by environmental aversive events can be assessed by different tests or protocols that require different degrees of time allocation, technical resources, and equipment. Rodent models of anhedonia are valuable tools in the study of the neurobiological mechanisms underpinning impaired behavioral responses and in the screening and characterization of drugs that may reverse these behavioral deficits. In particular, the antianhedonic or promotivational effects are relevant features in the spectrum of activities of drugs used in mood disorders or psychosis. Thus, more than the model, it is the choice of tests that is crucial since it influences which facets of anhedonia will be detected and should be tuned to the purpose of the study.
Collapse
Affiliation(s)
- Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena
| | | | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena,Correspondence: Carla Gambarana, Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 2 – 53100 Siena, Italy ()
| |
Collapse
|