1
|
Xu X, Song X, Chen F, Yan W, Meng Q, Liu J, Yao R, Liu Y, Dong F. Solifenacin promotes remyelination in cuprizone mouse model by inhibiting the Wnt/β-catenin signaling pathway. J Chem Neuroanat 2024; 136:102375. [PMID: 38123002 DOI: 10.1016/j.jchemneu.2023.102375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/22/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023]
Abstract
Demyelinating diseases are a type of neurological disorder characterized by the damage to the myelin sheath in the central nervous system. Promoting the proliferation and differentiation of oligodendrocyte precursor cells (OPCs) is crucial for treatment. Non-selective muscarinic receptor (MR) antagonists have been shown to improve remyelination in rodent models, although the mechanisms are still unclear. In this study, we treated cuprizone (CPZ)-induced demyelination mouse model with different concentrations of Solifenacin (Sol), a selective M3 receptor antagonist, to determine the optimal concentration for promoting remyelination. Behavioral tests and Luxol fast blue (LFB) staining were used to observe the extent of remyelination, while immunofluorescence was used to measure the expression levels of myelin-related proteins, including myelin basic protein (MBP) and platelet-derived growth factor receptor alpha (PDGFR-α). Western blot analysis was employed to analyze the expression levels of molecules associated with the Wnt/β-catenin signaling pathway. The results showed that Sol treatment significantly promoted myelin regeneration and OPCs differentiation in CPZ-induced demyelination mouse model. Additionally, Sol treatment inhibited the Wnt/β-catenin signaling pathway and reversed the effects of CPZ on OPCs differentiation. In conclusion, Sol may promote the differentiation of OPCs by inhibiting the Wnt/β-catenin signaling pathway, making it a potential therapeutic option for central nervous system demyelinating diseases.
Collapse
Affiliation(s)
- Xinqi Xu
- The First Clinical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Xueli Song
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Fei Chen
- The First Clinical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Weixing Yan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Qiqi Meng
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Jinfeng Liu
- School of Life Science, Xuzhou Medical University, Xuzhou, 221004, Jiangsu Province, China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Yaping Liu
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, 221004, Jiangsu Province, China
| | - Fuxing Dong
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China.
| |
Collapse
|
2
|
Xiang M, Zheng L, Pu D, Lin F, Ma X, Ye H, Pu D, Zhang Y, Wang D, Wang X, Zou K, Chen L, Zhang Y, Sun Z, Zhang T, Wu G. Intestinal Microbes in Patients With Schizophrenia Undergoing Short-Term Treatment: Core Species Identification Based on Co-Occurrence Networks and Regression Analysis. Front Microbiol 2022; 13:909729. [PMID: 35783418 PMCID: PMC9247572 DOI: 10.3389/fmicb.2022.909729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/12/2022] [Indexed: 01/12/2023] Open
Abstract
Schizophrenia, a common mental disorder, has a tremendous impact on the health and economy of people worldwide. Evidence suggests that the microbial-gut-brain axis is an important pathway for the interaction between the gut microbiome and the development of schizophrenia. What is not clear is how changes in the gut microbiota composition and structure during antipsychotic treatment improve the symptoms of schizophrenia. In this study, 25 patients with schizophrenia were recruited. Their fecal samples were collected before and after hospital treatment for 14–19 days. The composition and structure of the intestinal microbiota were evaluated by 16S rRNA sequencing analysis, and the results showed significant differences in fecal microbiota before and after treatment. Firmicutes (relative abundances of 82.60 and 86.64%) and Gemminger (relative abundances of 14.17 and 13.57%) were the first dominant species at the phylum and genus levels, respectively. The random forest algorithm and co-occurrence network analysis demonstrated that intestinal flora (especially the core species ASV57) could be used as biomarkers to distinguish different clinical states and match treatment regimens accordingly. In addition, after fecal microbiota transplantation, antibiotic-treated recipient mice showed multiple behavioral improvements. These included decreased psychomotor hyperactivity, increased social interaction, and memory. In conclusion, this study suggests that differences in the composition and structure of gut microbiota after treatment are associated with the development and severity of schizophrenia. Results may provide a potential target for the treatment of this disorder.
Collapse
Affiliation(s)
- Min Xiang
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Liqin Zheng
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Daoshen Pu
- The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Feng Lin
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Xiaodong Ma
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Huiqian Ye
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Daoqiong Pu
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Ying Zhang
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Dong Wang
- Psychiatry Department, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Xiaoli Wang
- Internal Medicine, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Kaiqing Zou
- The Outpatient Department, The Fourth People's Hospital of Ya'an, Ya'an, China
| | - Linqi Chen
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Zhang
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhanjiang Sun
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Zhang
- MOE Key Lab for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
- Tao Zhang
| | - Guolin Wu
- Medical Laboratory, The Fourth People's Hospital of Ya'an, Ya'an, China
- *Correspondence: Guolin Wu
| |
Collapse
|
3
|
What Can We Learn from Animal Models to Study Schizophrenia? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1400:15-33. [DOI: 10.1007/978-3-030-97182-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
4
|
Zhang N, Liu C, Zhang R, Jin L, Yin X, Zheng X, Siebert HC, Li Y, Wang Z, Loers G, Petridis AK. Amelioration of clinical course and demyelination in the cuprizone mouse model in relation to ketogenic diet. Food Funct 2021; 11:5647-5663. [PMID: 32539054 DOI: 10.1039/c9fo02944c] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ketogenic diet (KD) is defined as a high-fat, low-carbohydrate diet with appropriate amounts of protein, which has broad neuroprotective effects. However, the mechanisms of ameliorating the demyelination and of the neuroprotective effects of KD have not yet been completely elucidated. Therefore, the present study investigated the protection mechanism of KD treatment in the cuprizone (bis-cyclohexanone oxalydihydrazone, CPZ)-induced demyelination mice model, with special emphasis on neuroinflammation. After the KD treatment, an increased ketone body level in the blood of mice was detected, and a significant increase in the distance traveled within the central area was observed in the open field test, which reflected the increased exploration and decreased anxiety of mice that received CPZ. The results of Luxol fast blue and myelin basic protein (MBP) immunohistochemistry staining for the evaluation of the myelin content within the corpus callosum revealed a noticeable increase in the number of myelinated fibers and myelin score after KD administration in these animals. Concomitant, the protein expressions of glial fibrillary acidic protein (GFAP, an astrocyte marker), ionized calcium-binding adaptor molecule 1 (Iba-1, a microglial marker), CD68 (an activated microglia marker) and CD16/32 (a M1 microglial marker) were down-regulated, while the expression of oligodendrocyte lineage transcription factor 2 (OLIG2, an oligodendrocyte precursor cells marker) was up-regulated by the KD treatment. In addition, the KD treatment not only reduced the level of the C-X-C motif chemokine 10 (CXCL10), which is correlated to the recruitment of activated microglia, but also inhibited the production of proinflammatory cytokines, including interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α), which are closely correlated to the M1 phenotype microglia. It is noteworthy, that the expression levels of histone deacetylase 3 (HADC3) and nod-like receptor pyrin domain containing 3 (NLRP3) significantly decreased after KD administration. In conclusion, these data demonstrate that KD decreased the reactive astrocytes and activated the microglia in the corpus callosum, and that KD inhibited the HADC3 and NLRP3 inflammasome signaling pathway in CPZ-treated mice. This suggests that the inhibition of the HADC3 and NLRP3 signaling pathway may be a novel mechanism by which KD exerts its protective actions for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Chunhong Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Li Jin
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Xiaohan Yin
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Xuexing Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Hans-Christian Siebert
- RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Schauenburgerstr. 116, 24118 Kiel, Germany
| | - Yubao Li
- College of agriculture, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Falkenried 94, 20251 Hamburg, Germany
| | - Athanasios K Petridis
- Neurosurgical Department, Heinrich Heine University of Düsseldorf, Moorenstraße 5, 40255 Düsseldorf, Germany
| |
Collapse
|
5
|
Khodaei F, Khoshnoud MJ, Heidaryfar S, Heidari R, Karimpour Baseri MH, Azarpira N, Rashedinia M. The effect of ellagic acid on spinal cord and sciatica function in a mice model of multiple sclerosis. J Biochem Mol Toxicol 2020; 34:e22564. [PMID: 32640490 DOI: 10.1002/jbt.22564] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/08/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is a well-known neurodegenerative disorder, causing toxicity in different organs, such as spinal cord tissue. The goal of this study was to investigate the protective effect of ellagic acid (EA) against spinal cord and sciatica function in cuprizone (Cup)-induced demyelination model. Animals were divided into six equal groups. The first group received tap water as the control. Cup group was treated with Cup (0.2% w/w in fed). EA 100 group was orally treated with EA (100 mg/kg). EA + Cup groups were orally treated with three doses of 5, 50, and 100 mg/kg of EA plus Cup (0.2% w/w). All groups received treatment for 42 days. Open field, rotarod, and gait tests were done to evaluate the behavioral changes following Cup and/or EA treatment. Also, lipid peroxidation, reactive oxygen species (ROS) content, antioxidant capacity, superoxide dismutase (SOD), and catalase enzymes activity in spinal cord was evaluated. Luxol fast blue (LFB) staining also the behavioral tests were performed to evaluate the model. Cup increased ROS levels and oxidative stress in their spinal cord tissues. Also, Cup reduced antioxidant capacity, SOD, and catalase activity. EA (especially at 100 mg/kg) prevented these abnormal changes. EA co-treatment dose-dependently was able to ameliorate behavioral impairments in mice that received Cup. EA might act as a protective agent in MS by modulating spinal cord function.
Collapse
Affiliation(s)
- Forouzan Khodaei
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Mohammad Javad Khoshnoud
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Food and Supplements Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Heidaryfar
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad H Karimpour Baseri
- Department of Neuroscience and Addiction, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Rashedinia
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Yang L, Su Y, Guo F, Zhang H, Zhao Y, Huang Q, Xu H. Deep rTMS Mitigates Behavioral and Neuropathologic Anomalies in Cuprizone-Exposed Mice Through Reducing Microglial Proinflammatory Cytokines. Front Integr Neurosci 2020; 14:556839. [PMID: 33250722 PMCID: PMC7674917 DOI: 10.3389/fnint.2020.556839] [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] [Received: 04/29/2020] [Accepted: 10/05/2020] [Indexed: 02/05/2023] Open
Abstract
In comparison to conventional repetitive transcranial magnetic stimulation (rTMS), theta burst stimulation is stronger and more effective as a brain stimulation approach within short periods. Although this deep rTMS technique is being applied in treating neuropsychiatric disorders, few animal studies have attempted to clarify the neurobiological mechanisms underlying its beneficial effects. This animal study examined the effects of deep rTMS on the cuprizone-induced neuropathologic and behavioral anomalies and explored the underlying mechanism. Adolescent male C57BL/6 mice were fed a rodent chow without or with cuprizone (CPZ; 0.2% w/w) for 5 weeks. Another two groups of mice were subjected to deep rTMS or sham rTMS once a day during weeks 2-5 of the CPZ-feeding period. The behaviors of all mice were assessed after the withdrawal of CPZ before neuropathological and immunological analyses. Compared to the CNT group, mice in CPZ and CPZ + Sham groups showed deficits in social recognition and spatial working memory as well as anxiety-like behavior, in addition to myelin breakdown and OL loss in the corpus callosum (CC), caudate putamen, cerebral cortex, and hippocampus of the brain. Deep rTMS effectively reduced behavioral anomalies and blocked myelin breakdown and OL loss in CPZ-fed mice. Besides, it also dampened microglia activation at lesion sites and rectified cytokines levels (IL-1β, IL-6, and IL-10) in CPZ-affected regions. The most significant effect was seen in the cerebral cortex where alleviated neuropathology co-existed with less microglia activation and higher IL-10 level. These data provided experimental evidence for the beneficial effects of deep rTMS in CPZ-fed mice and revealed a neurobiological mechanism of the modality.
Collapse
Affiliation(s)
- Liu Yang
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Yawen Su
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Fannv Guo
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Handi Zhang
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Yinglin Zhao
- The Mental Health Center, Shantou University Medical College, Shantou, China
| | - Qinjun Huang
- The Mental Health Center, Shantou University Medical College, Shantou, China
- *Correspondence: Qinjun Huang Haiyun Xu
| | - Haiyun Xu
- The Mental Health Center, Shantou University Medical College, Shantou, China
- The School of Psychiatry, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qinjun Huang Haiyun Xu
| |
Collapse
|
7
|
Khodaei F, Rashedinia M, Heidari R, Rezaei M, Khoshnoud MJ. Ellagic acid improves muscle dysfunction in cuprizone-induced demyelinated mice via mitochondrial Sirt3 regulation. Life Sci 2019; 237:116954. [PMID: 31610192 DOI: 10.1016/j.lfs.2019.116954] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/24/2019] [Accepted: 10/10/2019] [Indexed: 01/10/2023]
Abstract
Sirt3 enzyme and mitochondrial abnormality can be related to excess fatigue or muscular dysfunction in multiple sclerosis (MS).Ellagic acid (EA) has a mitochondrial protector, iron chelator, antioxidant, and axon regenerator in neurons.In this study the effect of EAon muscle dysfunction, its mitochondria, and Sirt3 enzyme incuprizone-induced model of MSwas examined. Demyelination was induced by a diet containing 0.2% w/w cuprizone (Cup) for 42 days and EA administered daily (5, 50, and 100 mg/kg P.O) either with or without cuprizone in mice. Behavioral tests were assessed, and muscle tissue markers ofoxidative stress, mitochondrial parameters, mitochondrial respiratory chain activity, the Sirt3 protein level, and Sirt3 expression were also determined. Luxol fast blue staining and the behavioral tests were performed toassess the implemented model. In Cup group an increased oxidative stress in their muscle tissues was observed. Also, muscle mitochondria exhibited mitochondria dysfunction, lowered mitochondrial respiratory chain activity, Sirt3 protein level, and Sirt3 expression.EA prevented most of these anomalous alterations. Sub-chronicEA co-treatment dose-dependently ameliorated behavioral and muscular impairment in mice that received Cup.EA can effectively protect muscle tissue against cuprizone-induced demeylination via the mitochondrial protection, oxidative stress prevention and Sirt3 overexpression.
Collapse
Affiliation(s)
- Forouzan Khodaei
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Marzieh Rashedinia
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Rezaei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javad Khoshnoud
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Zahednasab H, Firouzi M, Kaboudanian-Ardestani S, Mojallal-Tabatabaei Z, Karampour S, Keyvani H. The protective effect of rifampicin on behavioral deficits, biochemical, and neuropathological changes in a cuprizone model of demyelination. Cytokine 2019; 113:417-426. [DOI: 10.1016/j.cyto.2018.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/16/2018] [Accepted: 10/18/2018] [Indexed: 12/19/2022]
|
9
|
Hershey JD, Gifford JJ, Zizza LJ, Pavlenko DA, Wagner GC, Miller S. Effects of Various Cleaning Agents on the Performance of Mice in Behavioral Assays of Anxiety. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2018; 57:335-339. [PMID: 29950249 DOI: 10.30802/aalas-jaalas-17-000161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cleaning behavioral equipment between rodent subjects is important to prevent disease transmission and reduce odor cues from previous subjects. However, the reporting regarding the cleansing procedures used during such experiments is sporadic and often incomplete. In addition, some investigators are reluctant to clean devices between subjects because they are concerned that animals will react negatively to the smell of the cleansing agents. We hypothesized that mice tested on an elevated plus maze (EPM) soiled with excretions from conspecifics would test as being more stressed than mice tested on the same apparatus that was cleaned between animals. We tested the performance of C57BL/6J mice on an EPM sanitized with 3 common cleaning agents-isopropyl alcohol, chlorine dioxide, and bleach-and on an EPM soiled with rodent urine, feces, and presumably pheromones. We further tested the potentially aversive nature of the cleansing agents by using the classic light:dark box and a 2-choice light:dark box. Our data indicate that cleaning the EPM compared with leaving it soiled did not affect performance in male or female C57 mice, nor did cleaning agent choice. In addition, test subjects did not react to the presence of the cleaning agents when incorporated into the classic light:dark test. However, in the 2-choice light:dark test, mice given the option to avoid an area containing a cleaning agent showed aversion to all 3 agents, when all other conditions were equal. Given the lack of an observable effect of cleaning on EPM performance, we recommend cleaning of the EPM device between C57 mice to minimize the potential spread of disease.
Collapse
Affiliation(s)
- John D Hershey
- Comparative Medicine Resources, Rutgers University, Piscataway, New Jersey;,
| | - Janace J Gifford
- Department of Psychology, Rutgers University, Piscataway, New Jersey
| | - Lauren J Zizza
- Office of Research Regulatory Affairs, Rutgers University, Piscataway, New Jersey
| | - Darya A Pavlenko
- Department of Psychology, Rutgers University, Piscataway, New Jersey
| | - George C Wagner
- Department of Psychology, Rutgers University, Piscataway, New Jersey
| | - Shoreh Miller
- Research Animal Facility, Rowan School of Medicine, Stratford, New Jersey
| |
Collapse
|