51
|
Suppressing aberrant phospholipase D1 signaling in 3xTg Alzheimer's disease mouse model promotes synaptic resilience. Sci Rep 2019; 9:18342. [PMID: 31797996 PMCID: PMC6892889 DOI: 10.1038/s41598-019-54974-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/21/2019] [Indexed: 02/08/2023] Open
Abstract
Current approaches in treatment of Alzheimer's disease (AD) is focused on early stages of cognitive decline. Identifying therapeutic targets that promote synaptic resilience during early stages may prevent progressive memory deficits by preserving memory mechanisms. We recently reported that the inducible isoform of phospholipase D (PLD1) was significantly increased in synaptosomes from post-mortem AD brains compared to age-matched controls. Using mouse models, we reported that the aberrantly elevated neuronal PLD1 is key for oligomeric amyloid driven synaptic dysfunction and underlying memory deficits. Here, we demonstrate that chronic inhibition using a well-tolerated PLD1 specific small molecule inhibitor is sufficient to prevent the progression of synaptic dysfunction during early stages in the 3xTg-AD mouse model. Firstly, we report prevention of cognitive decline in the inhibitor-treated group using novel object recognition (NOR) and fear conditioning (FC). Secondly, we provide electrophysiological assessment of better synaptic function in the inhibitor-treated group. Lastly, using Golgi staining, we report that preservation of dendritic spine integrity as one of the mechanisms underlying the action of the small molecule inhibitor. Collectively, these studies provide evidence for inhibition of PLD1 as a potential therapeutic strategy in preventing progression of cognitive decline associated with AD and related dementia.
Collapse
|
52
|
Wood RI, Serpa RO. Anabolic-androgenic steroid abuse and cognitive impairment: Testosterone IMPAIRS biconditional task performance in male rats. Behav Brain Res 2019; 379:112339. [PMID: 31697985 DOI: 10.1016/j.bbr.2019.112339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 01/01/2023]
Abstract
Our goal is to understand the consequences of anabolic-androgenic steroid (AAS) abuse on cognitive function, using rats as a model. There is relatively little research on how AAS abuse impacts cognition. In the present study, rats were tested for their ability to use contextual information to guide decision-making in biconditional discrimination. The Stroop task is a classic human test for contextual decision-making. In rodents, biconditional discrimination challenges subjects to use contextual cues in the operant chamber to resolve the correct lever response when auditory and visual cues are incongruent. The hypothesis is that chronic high-dose testosterone impairs biconditional discrimination. Rats were trained in 24 trials/day over 14 days, in alternating sessions with each environment. On a flat floor with houselight illuminated, auditory cues (clicker vs tone) signified the active lever. On a barred floor with no light, visual cues from 2 stimulus lights (constant vs blinking) identified the active lever. Rats treated chronically with testosterone (7.5 mg/kg) were unimpaired in task acquisition, and all rats learned to select the correct lever in response to auditory or visual cues. During extinction, controls made significantly more correct than incorrect responses in congruent trials (p < 0.05 by paired t-test), but testosterone-treated rats failed to show a similar preference. This was reflected by significant interactions of drug x cue agreement (F1,18 = 5.21, p < 0.05) and drug x cue agreement x response accuracy (F1,18 = 8.95, p < 0.05). These results suggest that testosterone impairs cognitive flexibility, and demonstrates potential for AAS abuse to impair cognitive function in humans.
Collapse
Affiliation(s)
- Ruth I Wood
- Department of Integrative Anatomical Sciences, Keck School of Medicine at the University of Southern California, Los Angeles, CA, 90033, United States.
| | - Rebecka O Serpa
- Department of Integrative Anatomical Sciences, Keck School of Medicine at the University of Southern California, Los Angeles, CA, 90033, United States
| |
Collapse
|
53
|
Stefanello FV, Fontana BD, Ziani PR, Müller TE, Mezzomo NJ, Rosemberg DB. Exploring Object Discrimination in Zebrafish: Behavioral Performance and Scopolamine-Induced Cognitive Deficits at Different Retention Intervals. Zebrafish 2019; 16:370-378. [DOI: 10.1089/zeb.2018.1703] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Flavia V. Stefanello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Barbara D. Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Paola R. Ziani
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Talise E. Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Nathana J. Mezzomo
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Denis B. Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, Louisiana
| |
Collapse
|
54
|
Effects of Single Cage Housing on Stress, Cognitive, and Seizure Parameters in the Rat and Mouse Pilocarpine Models of Epilepsy. eNeuro 2019; 6:ENEURO.0179-18.2019. [PMID: 31331937 PMCID: PMC6709207 DOI: 10.1523/eneuro.0179-18.2019] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/08/2019] [Accepted: 05/29/2019] [Indexed: 01/01/2023] Open
Abstract
Many experimental approaches require housing rodents in individual cages, including in epilepsy research. However, rats and mice are social animals; and individual housing constitutes a stressful situation. The goal of the present study was to determine the effects of individual housing as compared to conditions maintaining social contact on stress markers and epilepsy. Control male mice socially housed during pretest and then transferred to individual cages for six weeks displayed anhedonia, increased anxiety and biological markers of stress as compared to pretest values or mice kept socially housed during six weeks. Pilocarpine (pilo)-treated mice housed together showed increased levels of anhedonia, anxiety and stress markers as well as decreased cognitive performance as compared to the control group. The differences were more significant in pilo-treated mice housed individually. Anxiety correlated linearly with cognitive performance and stress markers independently of the experimental conditions. In the male rat pilo model, seizures were sixteen times more frequent in singly housed animals as compared to animals kept in pairs. Daily interactions with an experimenter in otherwise singly housed animals was sufficient to produce results identical to those found in animals kept in pairs. We propose that social isolation produces a severe phenotype in terms of stress and seizure frequency as compared to animals maintaining social contact (at least in these two models), a factor that needs to be taken into account for data interpretation, in particular for preclinical studies.
Collapse
|
55
|
Schober ME, Requena DF, Casper TC, Velhorst AK, Lolofie A, McFarlane KE, Otto TE, Terry C, Gensel JC. Docosahexaenoic acid decreased neuroinflammation in rat pups after controlled cortical impact. Exp Neurol 2019; 320:112971. [PMID: 31247195 DOI: 10.1016/j.expneurol.2019.112971] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 05/27/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) is the leading cause of acquired neurologic disability in children, yet specific therapies to treat TBI are lacking. Therapies that decrease the inflammatory response and enhance a reparative immune action may decrease oxidative damage and improve outcomes after TBI. Docosahexaenoic acid (DHA) modulates the immune response to injury in many organs. DHA given in the diet before injury decreased rat pup cognitive impairment, oxidative stress and white matter injury in our developmental TBI model using controlled cortical impact (CCI). Little is known about DHA effects on neuroinflammation in the developing brain. Further, it is not known if DHA given after developmental TBI exerts neuroprotective effects. We hypothesized that acute DHA treatment would decrease oxidative stress and improve cognitive outcome, associated with decreased pro-inflammatory activation of microglia, the brain's resident macrophages. METHODS 17-day-old rat pups received intraperitoneal DHA or vehicle after CCI or SHAM surgery followed by DHA diet or continuation of REG diet to create DHACCI, REGCCI, SHAMDHA and SHAMREG groups. We measured brain nitrates/nitrites (NOx) at post injury day (PID) 1 to assess oxidative stress. We tested memory using Novel Object Recognition (NOR) at PID14. At PID 3 and 7, we measured reactivity of microglial activation markers Iba1, CD68 and CD206 and astrocyte marker GFAP in the injured cortex. At PID3, 7 and 30 we measured mRNA levels of inflammation-related genes and transcription factors in flow-sorted brain cells. RESULTS DHA decreased oxidative stress at PID1 and pro-inflammatory microglial activation at PID3. CCI increased mRNA levels of two interferon regulatory family transcription factors, blunted by DHA, particularly in microglia-enriched cell populations at PID7. CCI increased mRNA levels of genes associated with "pro- " and "anti-" inflammatory activity at PID3, 7 and 30. Most notably within the microglia-enriched population, DHA blunted increased mRNA levels of pro-inflammatory genes at PID 3 and 7 and of anti-inflammatory genes at PID 30. Particularly in microglia, we observed parallel activation of pro-inflammatory and anti-inflammatory genes. DHA improved performance on NOR at PID14 after CCI. CONCLUSIONS DHA decreased oxidative stress and histologic and mRNA markers of microglial pro-inflammatory activation in rat pup brain acutely after CCI associated with improved short term cognitive function. DHA administration after CCI has neuroprotective effects, which may result in part from modulation of microglial activation toward a less inflammatory profile in the first week after CCI. Future and ongoing studies will focus on phagocytic function and reactive oxygen species production in microglia and macrophages to test functional effects of DHA on neuroinflammation in our model. Given its favorable safety profile in children, DHA is a promising candidate therapy for pediatric TBI.
Collapse
Affiliation(s)
- Michelle E Schober
- Department of Pediatrics, Division of Critical Care University of Utah, Salt Lake City, UT 84132, United States.
| | - Daniela F Requena
- Department of Pediatrics, Division of Critical Care University of Utah, Salt Lake City, UT 84132, United States
| | - T Charles Casper
- Department of Pediatrics, Division of Critical Care University of Utah, Salt Lake City, UT 84132, United States.
| | - Amy K Velhorst
- Department of Physiology and Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Alyssa Lolofie
- Department of Pediatrics, Division of Critical Care University of Utah, Salt Lake City, UT 84132, United States.
| | - Katelyn E McFarlane
- Department of Physiology and Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| | - Taylor E Otto
- Department of Physiology and Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Cynthia Terry
- Department of Pediatrics, Division of Critical Care University of Utah, Salt Lake City, UT 84132, United States.
| | - John C Gensel
- Department of Physiology and Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| |
Collapse
|
56
|
Marechal D, Brault V, Leon A, Martin D, Lopes Pereira P, Loaëc N, Birling MC, Friocourt G, Blondel M, Herault Y. Cbs overdosage is necessary and sufficient to induce cognitive phenotypes in mouse models of Down syndrome and interacts genetically with Dyrk1a. Hum Mol Genet 2019; 28:1561-1577. [PMID: 30649339 DOI: 10.1093/hmg/ddy447] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 01/16/2023] Open
Abstract
Identifying dosage-sensitive genes is a key to understand the mechanisms underlying intellectual disability in Down syndrome (DS). The Dp(17Abcg1-Cbs)1Yah DS mouse model (Dp1Yah) shows cognitive phenotypes that need to be investigated to identify the main genetic driver. Here, we report that three copies of the cystathionine-beta-synthase gene (Cbs) in the Dp1Yah mice are necessary to observe a deficit in the novel object recognition (NOR) paradigm. Moreover, the overexpression of Cbs alone is sufficient to induce deficits in the NOR test. Accordingly, overexpressing human CBS specifically in Camk2a-expressing neurons leads to impaired objects discrimination. Altogether, this shows that Cbs overdosage is involved in DS learning and memory phenotypes. To go further, we identified compounds that interfere with the phenotypical consequence of CBS overdosage in yeast. Pharmacological intervention in Tg(CBS) mice with one selected compound restored memory in the NOR test. In addition, using a genetic approach, we demonstrated an epistatic interaction between Cbs and Dyrk1a, another human chromosome 21-located gene (which encodes the dual-specificity tyrosine phosphorylation-regulated kinase 1a) and an already known target for DS therapeutic intervention. Further analysis using proteomic approaches highlighted several molecular pathways, including synaptic transmission, cell projection morphogenesis and actin cytoskeleton, that are affected by DYRK1A and CBS overexpression. Overall, we demonstrated that CBS overdosage underpins the DS-related recognition memory deficit and that both CBS and DYRK1A interact to control accurate memory processes in DS. In addition, our study establishes CBS as an intervention point for treating intellectual deficiencies linked to DS.
Collapse
Affiliation(s)
- Damien Marechal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Véronique Brault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Alice Leon
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Dehren Martin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Patricia Lopes Pereira
- Transgenese et Archivage Animaux Modèles, TAAM, CNRS, 3B Rue de la Férollerie Orléans, France
| | - Nadege Loaëc
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | | | - Gaelle Friocourt
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Marc Blondel
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Illkirch, France
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris, ICS, Illkirch, France
| |
Collapse
|
57
|
Moreton E, Baron P, Tiplady S, McCall S, Clifford B, Langley-Evans S, Fone K, Voigt J. Impact of early exposure to a cafeteria diet on prefrontal cortex monoamines and novel object recognition in adolescent rats. Behav Brain Res 2019; 363:191-198. [DOI: 10.1016/j.bbr.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/24/2019] [Accepted: 02/02/2019] [Indexed: 12/19/2022]
|
58
|
Ryu T, Park HJ, Kim H, Cho YC, Kim BC, Jo J, Seo YW, Choi WS, Kim K. Improved memory and reduced anxiety in δ-catenin transgenic mice. Exp Neurol 2019; 318:22-31. [PMID: 30981806 DOI: 10.1016/j.expneurol.2019.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 03/13/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022]
Abstract
δ-Catenin is abundant in the brain and affects its synaptic plasticity. Furthermore, loss of δ-catenin is related to the deficits of learning and memory, mental retardation (cri-du-chat syndrome), and autism. A few studies about δ-catenin deficiency mice were performed. However, the effect of δ-catenin overexpression in the brain has not been investigated as yet. Therefore we generated a δ-catenin overexpressing mouse model. To generate a transgenic mouse model overexpressing δ-catenin in the brain, δ-catenin plasmid having a Thy-1 promotor was microinjected in C57BL/6 mice. Our results showed δ-catenin transgenic mice expressed higher levels of N-cadherin, β-catenin, and p120-catenin than did wild type mice. Furthermore, δ-catenin transgenic mice exhibited better object recognition, better sociability, and lower anxiety than wild type mice. However, both mice groups showed a similar pattern in locomotion tests. Although δ-catenin transgenic mice show similar locomotion, they show improved sociability and reduced anxiety. These characteristics are opposite to the symptoms of autism or mental retardation, which are caused when δ-catenin is deficient. These results suggest that δ-catenin may alleviate symptoms of autism, Alzheimer's disease and mental retardation.
Collapse
Affiliation(s)
- Taeyong Ryu
- College of Pharmacy and Research Institute for Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyung Joon Park
- School of Biological Sciences and Technology, College of Natural Sciences, College of Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hangun Kim
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 57922, Republic of Korea
| | - Young-Chang Cho
- College of Pharmacy and Research Institute for Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Medical School, Gwnagju 61469, Republic of Korea
| | - Jihoon Jo
- Department of Neurology, Chonnam National University Medical School, Gwnagju 61469, Republic of Korea
| | - Young-Woo Seo
- Korea Basic Science Institute, Gwangju Center, Gwangju 61186, Republic of Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, College of Natural Sciences, College of Medicine, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Kwonseop Kim
- College of Pharmacy and Research Institute for Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
59
|
Bruinenberg VM, van Vliet D, van der Goot E, Counotte DS, Kuhn M, van Spronsen FJ, van der Zee EA. Long-term dietary intervention with low Phe and/or a specific nutrient combination improve certain aspects of brain functioning in phenylketonuria (PKU). PLoS One 2019; 14:e0213391. [PMID: 30875376 PMCID: PMC6420157 DOI: 10.1371/journal.pone.0213391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/19/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION In phenylketonuria (PKU), a gene mutation in the phenylalanine metabolic pathway causes accumulation of phenylalanine (Phe) in blood and brain. Although early introduction of a Phe-restricted diet can prevent severe symptoms from developing, patients who are diagnosed and treated early still experience deficits in cognitive functioning indicating shortcomings of current treatment. In the search for new and/or additional treatment strategies, a specific nutrient combination (SNC) was postulated to improve brain function in PKU. In this study, a long-term dietary intervention with a low-Phe diet, a specific combination of nutrients designed to improve brain function, or both concepts together was investigated in male and female BTBR PKU and WT mice. MATERIAL & METHODS 48 homozygous wild-types (WT, +/+) and 96 PKU BTBRPah2 (-/-) male and female mice received dietary interventions from postnatal day 31 till 10 months of age and were distributed in the following six groups: high Phe diet (WT C-HP, PKU C-HP), high Phe plus specific nutrient combination (WT SNC-HP, PKU SNC-HP), PKU low-Phe diet (PKU C-LP), and PKU low-Phe diet plus specific nutrient combination (PKU SNC- LP). Memory and motor function were tested at time points 3, 6, and 9 months after treatment initiation in the open field (OF), novel object recognition test (NOR), spatial object recognition test (SOR), and the balance beam (BB). At the end of the experiments, brain neurotransmitter concentrations were determined. RESULTS In the NOR, we found that PKU mice, despite being subjected to high Phe conditions, could master the task on all three time points when supplemented with SNC. Under low Phe conditions, PKU mice on control diet could master the NOR at all three time points, while PKU mice on the SNC supplemented diet could master the task at time points 6 and 9 months. SNC supplementation did not consistently influence the performance in the OF, SOR or BB in PKU mice. The low Phe diet was able to normalize concentrations of norepinephrine and serotonin; however, these neurotransmitters were not influenced by SNC supplementation. CONCLUSION This study demonstrates that both a long-lasting low Phe diet, the diet enriched with SNC, as well as the combined diet was able to ameliorate some, but not all of these PKU-induced abnormalities. Specifically, this study is the first long-term intervention study in BTBR PKU mice that shows that SNC supplementation can specifically improve novel object recognition.
Collapse
Affiliation(s)
- Vibeke M. Bruinenberg
- Molecular Neurobiology, GELIFES, University of Groningen, Groningen, The Netherlands
| | - Danique van Vliet
- Division of Metabolic Diseases, Beatrix Children’s Hospital, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| | - Els van der Goot
- Molecular Neurobiology, GELIFES, University of Groningen, Groningen, The Netherlands
| | | | | | - Francjan J. van Spronsen
- Division of Metabolic Diseases, Beatrix Children’s Hospital, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| | - Eddy A. van der Zee
- Molecular Neurobiology, GELIFES, University of Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
60
|
Souchet B, Duchon A, Gu Y, Dairou J, Chevalier C, Daubigney F, Nalesso V, Créau N, Yu Y, Janel N, Herault Y, Delabar JM. Prenatal treatment with EGCG enriched green tea extract rescues GAD67 related developmental and cognitive defects in Down syndrome mouse models. Sci Rep 2019; 9:3914. [PMID: 30850713 PMCID: PMC6408590 DOI: 10.1038/s41598-019-40328-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 01/31/2019] [Indexed: 12/21/2022] Open
Abstract
Down syndrome is a common genetic disorder caused by trisomy of chromosome 21. Brain development in affected foetuses might be improved through prenatal treatment. One potential target is DYRK1A, a multifunctional kinase encoded by chromosome 21 that, when overexpressed, alters neuronal excitation-inhibition balance and increases GAD67 interneuron density. We used a green tea extract enriched in EGCG to inhibit DYRK1A function only during gestation of transgenic mice overexpressing Dyrk1a (mBACtgDyrk1a). Adult mice treated prenatally displayed reduced levels of inhibitory markers, restored VGAT1/VGLUT1 balance, and rescued density of GAD67 interneurons. Similar results for gabaergic and glutamatergic markers and interneuron density were obtained in Dp(16)1Yey mice, trisomic for 140 chromosome 21 orthologs; thus, prenatal EGCG exhibits efficacy in a more complex DS model. Finally, cognitive and behaviour testing showed that adult Dp(16)1Yey mice treated prenatally had improved novel object recognition memory but do not show improvement with Y maze paradigm. These findings provide empirical support for a prenatal intervention that targets specific neural circuitries.
Collapse
Affiliation(s)
- Benoit Souchet
- Université Paris-Diderot, Sorbonne Paris Cité, Adaptive Functional Biology, National Centre for Scientific Research (CNRS), UMR 8251, Paris, France
| | - Arnaud Duchon
- Institut Génétique Biologie Moléculaire Cellulaire, CNRS, French National Institute of Health and Medical Research (INSERM), UMR 7104, UMR 964, Illkirch, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
- CNRS, UMR 7104, Illkirch, France
- INSERM, U964, Illkirch, France
- Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
| | - Yuchen Gu
- Université Paris-Diderot, Sorbonne Paris Cité, Adaptive Functional Biology, National Centre for Scientific Research (CNRS), UMR 8251, Paris, France
| | - Julien Dairou
- CNRS, UMR 8601, Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Descartes-Sorbonne Paris Cité, 75270, Paris, France
| | - Claire Chevalier
- Institut Génétique Biologie Moléculaire Cellulaire, CNRS, French National Institute of Health and Medical Research (INSERM), UMR 7104, UMR 964, Illkirch, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
- CNRS, UMR 7104, Illkirch, France
- INSERM, U964, Illkirch, France
- Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
| | - Fabrice Daubigney
- Université Paris-Diderot, Sorbonne Paris Cité, Adaptive Functional Biology, National Centre for Scientific Research (CNRS), UMR 8251, Paris, France
| | - Valérie Nalesso
- Institut Génétique Biologie Moléculaire Cellulaire, CNRS, French National Institute of Health and Medical Research (INSERM), UMR 7104, UMR 964, Illkirch, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
- CNRS, UMR 7104, Illkirch, France
- INSERM, U964, Illkirch, France
- Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France
| | - Nicole Créau
- Université Paris-Diderot, Sorbonne Paris Cité, Adaptive Functional Biology, National Centre for Scientific Research (CNRS), UMR 8251, Paris, France
| | - Yuejin Yu
- Children's Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Nathalie Janel
- Université Paris-Diderot, Sorbonne Paris Cité, Adaptive Functional Biology, National Centre for Scientific Research (CNRS), UMR 8251, Paris, France
- Children's Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Yann Herault
- Institut Génétique Biologie Moléculaire Cellulaire, CNRS, French National Institute of Health and Medical Research (INSERM), UMR 7104, UMR 964, Illkirch, France.
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.
- CNRS, UMR 7104, Illkirch, France.
- INSERM, U964, Illkirch, France.
- Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.
| | - Jean Maurice Delabar
- Université Paris-Diderot, Sorbonne Paris Cité, Adaptive Functional Biology, National Centre for Scientific Research (CNRS), UMR 8251, Paris, France.
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICM, Paris, France.
- Brain & Spine Institute (ICM) CNRS UMR7225, Inserm UMRS 975, Paris, France.
| |
Collapse
|
61
|
Schaefer ML, Wang M, Perez PJ, Coca Peralta W, Xu J, Johns RA. Nitric Oxide Donor Prevents Neonatal Isoflurane-induced Impairments in Synaptic Plasticity and Memory. Anesthesiology 2019; 130:247-262. [PMID: 30601214 PMCID: PMC6538043 DOI: 10.1097/aln.0000000000002529] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
WHAT WE ALREADY KNOW ABOUT THIS TOPIC Some general anesthetics have been shown to have adverse effects on neuronal development that affect neural function and cognitive behavior.Clinically relevant concentrations of inhalational anesthetics inhibit the postsynaptic density (PSD)-95, discs large homolog, and zona occludens-1 (PDZ) domain-mediated protein-protein interaction between PSD-95 or PSD-93 and N-methyl-D-aspartate receptors or neuronal NO synthase. WHAT THIS ARTICLE TELLS US THAT IS NEW Neonatal PSD-95 PDZ2WT peptide treatment mimics the effects of isoflurane (~1 minimum alveolar concentration) by altering dendritic spine morphology, neural plasticity, and memory without inducing detectable increases in apoptosis or changes in synaptic density.These results indicate that a single dose of isoflurane (~1 minimum alveolar concentration) or PSD-95 PDZ2WT peptide alters dendritic spine architecture and functions important for cognition in the developing brain. This impairment can be prevented by administration of the NO donor molsidomine. BACKGROUND In humans, multiple early exposures to procedures requiring anesthesia constitute a significant risk factor for development of learning disabilities and disorders of attention. In animal studies, newborns exposed to anesthetics develop long-term deficits in cognition. Previously, our laboratory showed that postsynaptic density (PSD)-95, discs large homolog, and zona occludens-1 (PDZ) domains may serve as a molecular target for inhaled anesthetics. This study investigated a role for PDZ interactions in spine development, plasticity, and memory as a potential mechanism for early anesthetic exposure-produced cognitive impairment. METHODS Postnatal day 7 mice were exposed to 1.5% isoflurane for 4 h or injected with 8 mg/kg active PSD-95 PDZ2WT peptide. Apoptosis, hippocampal dendritic spine changes, synapse density, long-term potentiation, and cognition functions were evaluated (n = 4 to 18). RESULTS Exposure of postnatal day 7 mice to isoflurane or PSD-95 PDZ2WT peptide causes a reduction in long thin spines (median, interquartile range [IQR]: wild type control [0.54, 0.52 to 0.86] vs. wild type isoflurane [0.31, 0.16 to 0.38], P = 0.034 and PDZ2MUT [0.86, 0.67 to 1.0] vs. PDZ2WT [0.55, 0.53 to 0.59], P = 0.028), impairment in long-term potentiation (median, IQR: wild type control [123, 119 to 147] and wild type isoflurane [101, 96 to 118], P = 0.049 and PDZ2MUT [125, 119 to 131] and PDZ2WT [104, 97 to 107], P = 0.029), and deficits in acute object recognition (median, IQR: wild type control [79, 72 to 88] vs. wild type isoflurane [63, 55 to 72], P = 0.044 and PDZ2MUT [81, 69 to 84] vs. PDZ2WT [67, 57 to 77], P = 0.039) at postnatal day 21 without inducing detectable differences in apoptosis or changes in synaptic density. Impairments in recognition memory and long-term potentiation were preventable by introduction of a NO donor. CONCLUSIONS Early disruption of PDZ domain-mediated protein-protein interactions alters spine morphology, synaptic function, and memory. These results support a role for PDZ interactions in early anesthetic exposure-produced cognitive impairment. Prevention of recognition memory and long-term potentiation deficits with a NO donor supports a role for the N-methyl-D-aspartate receptor/PSD-95/neuronal NO synthase pathway in mediating these aspects of isoflurane-induced cognitive impairment.
Collapse
Affiliation(s)
- Michele L Schaefer
- From the Department Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
62
|
PSD95 and nNOS interaction as a novel molecular target to modulate conditioned fear: relevance to PTSD. Transl Psychiatry 2018; 8:155. [PMID: 30108200 PMCID: PMC6092346 DOI: 10.1038/s41398-018-0208-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/10/2018] [Indexed: 11/21/2022] Open
Abstract
Stimulation of N-methyl-D-aspartic acid receptors (NMDARs) and the resulting increase of nitric oxide (NO) production are critical for fear memory formation. Following NMDAR activation, efficient production of NO requires linking the 95 kDa postsynaptic density protein (PSD95), a scaffolding protein to neuronal nitric oxide synthase (nNOS). A variety of previously studied NMDAR antagonists and NOS inhibitors can disrupt fear conditioning, but they also affect many other CNS functions such as motor activity, anxiety, and learning. We hypothesized that disrupting nNOS and PSD95 interaction in the amygdala, a critical site for fear memory formation, will reduce conditioned fear. Our results show that systemic treatment with ZL006, a compound that disrupts PSD95/nNOS binding, attenuates fear memory compared to its inactive isomer ZL007. Co-immunoprecipitation after fear conditioning showed a robust increase in the amygdala PSD95/nNOS binding, which was blocked by systemic pre-administration of ZL006. Treatment of amygdala slices with ZL006 also impaired long-term potentiation (LTP), a cellular signature of synaptic plasticity. Direct intra-amygdala infusion of ZL006 also attenuated conditioned fear. Finally, unlike NMDAR antagonist MK-801, ZL006 does not affect locomotion, social interaction, object recognition memory, and spatial memory. These findings support the hypothesis that disrupting the PSD95/nNOS interaction downstream of NMDARs selectively reduces fear memory, and highlights PSD95/nNOS interaction as a novel target for fear-related disorders, such as posttraumatic stress disorder.
Collapse
|
63
|
Schober ME, Requena DF, Rodesch CK. EPO improved neurologic outcome in rat pups late after traumatic brain injury. Brain Dev 2018; 40:367-375. [PMID: 29429559 PMCID: PMC5878733 DOI: 10.1016/j.braindev.2018.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/11/2017] [Accepted: 01/12/2018] [Indexed: 11/25/2022]
Abstract
UNLABELLED In adult rats, erythropoietin improved outcomes early and late after traumatic brain injury, associated with increased levels of Brain Derived Neurotrophic Factor. Using our model of pediatric traumatic brain injury, controlled cortical impact in 17-day old rats, we previously showed that erythropoietin increased hippocampal neuronal fraction in the first two days after injury. Erythropoietin also decreased activation of caspase3, an apoptotic enzyme modulated by Brain Derived Neurotrophic Factor, and improved Novel Object Recognition testing 14 days after injury. Data on long-term effects of erythropoietin on Brain Derived Neurotrophic Factor expression, histology and cognitive function after developmental traumatic brain injury are lacking. We hypothesized that erythropoietin would increase Brain Derived Neurotrophic Factor and improve long-term object recognition in rat pups after controlled cortical impact, associated with increased neuronal fraction in the hippocampus. METHODS Rats pups received erythropoietin or vehicle at 1, 24, and 48 h and 7 days after injury or sham surgery followed by histology at 35 days, Novel Object Recognition testing at adulthood, and Brain Derived Neurotrophic Factor measurements early and late after injury. RESULTS Erythropoietin improved Novel Object Recognition performance and preserved hippocampal volume, but not neuronal fraction, late after injury. CONCLUSIONS Improved object recognition in erythropoietin treated rats was associated with preserved hippocampal volume late after traumatic brain injury. Erythropoietin is approved to treat various pediatric conditions. Coupled with exciting experimental and clinical studies suggesting it is beneficial after neonatal hypoxic ischemic brain injury, our preliminary findings support further study of erythropoietin use after developmental traumatic brain injury.
Collapse
Affiliation(s)
- Michelle E Schober
- Department of Pediatrics, Division of Critical Care, University of Utah, Salt Lake City, UT 84132, United States.
| | - Daniela F Requena
- Department of Pediatrics, Division of Critical Care, University of Utah, Salt Lake City, UT, United States 84132
| | - Christopher K Rodesch
- Core Facilities and Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, United States 84132
| |
Collapse
|
64
|
Templer VL, Wise TB, Dayaw KIT, Dayaw JNT. Nonsocially housed rats (Ratus norvegicus) seek social interactions and social novelty more than socially housed counterparts. ACTA ACUST UNITED AC 2018; 132:240-252. [PMID: 29683686 DOI: 10.1037/com0000112] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sociability is the act or quality of social interaction and can be quantified by determining the number and duration of interactions with conspecifics. The purpose of this study was to examine the extent to which sustained social contact, as achieved by constant social living conditions, influenced social behavior. Beginning in juvenility, 19 male Long-Evans rats were housed in enriched environments, with half living socially in a large group and half living individually. After several months in these housing conditions, rats were tested on a sociality test and a social novelty preference test. Nonsocially housed rats exhibited more social behavior than socially housed rats. In the sociality test, nonsocially housed rats engaged with an unfamiliar rat more than socially housed rats. Similarly, in the social novelty test, nonsocially housed rats visited a novel stranger more than the now-familiar rat (from the sociality test) as compared with the socially housed rats. It is unlikely that general anxiety factors can account for between-groups social effects, as there were no group differences in behavior on the elevated zero maze and open field test. Furthermore, socially and nonsocially housed rats were matched in spontaneous object exploration and novelty preference in a novel object recognition test, eliminating the possibility that general exploratory behavior or novelty preference accounted for group differences in the sociability tasks. These results suggest that lack of social interaction in nonsocially housed rats may be more powerful for social motivation than the consistent opportunity for social contact afforded by social living conditions. (PsycINFO Database Record
Collapse
|
65
|
Grégoire CA, Tobin S, Goldenstein BL, Samarut É, Leclerc A, Aumont A, Drapeau P, Fulton S, Fernandes KJL. RNA-Sequencing Reveals Unique Transcriptional Signatures of Running and Running-Independent Environmental Enrichment in the Adult Mouse Dentate Gyrus. Front Mol Neurosci 2018; 11:126. [PMID: 29706867 PMCID: PMC5908890 DOI: 10.3389/fnmol.2018.00126] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/29/2018] [Indexed: 11/18/2022] Open
Abstract
Environmental enrichment (EE) is a powerful stimulus of brain plasticity and is among the most accessible treatment options for brain disease. In rodents, EE is modeled using multi-factorial environments that include running, social interactions, and/or complex surroundings. Here, we show that running and running-independent EE differentially affect the hippocampal dentate gyrus (DG), a brain region critical for learning and memory. Outbred male CD1 mice housed individually with a voluntary running disk showed improved spatial memory in the radial arm maze compared to individually- or socially-housed mice with a locked disk. We therefore used RNA sequencing to perform an unbiased interrogation of DG gene expression in mice exposed to either a voluntary running disk (RUN), a locked disk (LD), or a locked disk plus social enrichment and tunnels [i.e., a running-independent complex environment (CE)]. RNA sequencing revealed that RUN and CE mice showed distinct, non-overlapping patterns of transcriptomic changes versus the LD control. Bio-informatics uncovered that the RUN and CE environments modulate separate transcriptional networks, biological processes, cellular compartments and molecular pathways, with RUN preferentially regulating synaptic and growth-related pathways and CE altering extracellular matrix-related functions. Within the RUN group, high-distance runners also showed selective stress pathway alterations that correlated with a drastic decline in overall transcriptional changes, suggesting that excess running causes a stress-induced suppression of running’s genetic effects. Our findings reveal stimulus-dependent transcriptional signatures of EE on the DG, and provide a resource for generating unbiased, data-driven hypotheses for novel mediators of EE-induced cognitive changes.
Collapse
Affiliation(s)
- Catherine-Alexandra Grégoire
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada.,CNS Research Group, University of Montreal, Montreal, QC, Canada.,Department of Pathology and Cell Biology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Stephanie Tobin
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada.,Department of Nutrition, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Brianna L Goldenstein
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada.,CNS Research Group, University of Montreal, Montreal, QC, Canada.,Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Éric Samarut
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada.,CNS Research Group, University of Montreal, Montreal, QC, Canada.,Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Andréanne Leclerc
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Anne Aumont
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada
| | - Pierre Drapeau
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada.,CNS Research Group, University of Montreal, Montreal, QC, Canada.,Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Stephanie Fulton
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada.,Department of Nutrition, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Karl J L Fernandes
- Research Center of the University of Montreal Hospital, University of Montreal, Montreal, QC, Canada.,CNS Research Group, University of Montreal, Montreal, QC, Canada.,Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
66
|
Sargolzaei S, Cai Y, Lee D, Harris NG, Giza CC. Quantification of Biological Responses as Predictors of Cognitive Outcome after Developmental TBI. ... IEEE-EMBS INTERNATIONAL CONFERENCE ON BIOMEDICAL AND HEALTH INFORMATICS. IEEE-EMBS INTERNATIONAL CONFERENCE ON BIOMEDICAL AND HEALTH INFORMATICS 2018; 2018:381-384. [PMID: 34337425 DOI: 10.1109/bhi.2018.8333448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Successful translational studies within the field of Traumatic Brain Injury (TBI) are concerned with determining reliable markers of injury outcome at chronic time points. Determination of injury severity following Fluid Percussion Injury (FPI) has long been limited to the measured atmospheric pressure associated with the delivered pulse. Duration of unresponsiveness to toe pinch (unconsciousness) was next introduced as an extra marker of injury severity. The current study is an effort to assess the utilization of acute injury-induced biological responses (duration of toe pinch unresponsiveness, percent body weight change, quantification of brain edema, and apnea duration) to predict cognitive performance at a subacute time point following developmental brain injury. Cognitive performance, when measured at a subacute phase, after developmental FPI was negatively correlated with the following variables, duration of toe pinch unresponsiveness, percent weight change, and quantified level of brain edema. These finding suggest the potential utilization of reliable severity assessment of injury-induced biological responses in determining outcome measures at subacute time points.
Collapse
Affiliation(s)
- Saman Sargolzaei
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Yan Cai
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Deborah Lee
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Neil G Harris
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Christopher C Giza
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
67
|
Travaglia A, Steinmetz AB, Miranda JM, Alberini CM. Mechanisms of critical period in the hippocampus underlie object location learning and memory in infant rats. Learn Mem 2018; 25:176-182. [PMID: 29545389 PMCID: PMC5855526 DOI: 10.1101/lm.046946.117] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/12/2018] [Indexed: 12/29/2022]
Abstract
Episodic memories in early childhood are rapidly forgotten, a phenomenon that is associated with "infantile amnesia," the inability of adults to remember early-life experiences. We recently showed that early aversive contextual memory in infant rats, which is in fact rapidly forgotten, is actually not lost, as reminders presented later in life reinstate a long-lasting and context-specific memory. We also showed that the formation of this infantile memory recruits in the hippocampus mechanisms typical of developmental critical periods. Here, we tested whether similar mechanisms apply to a nonaversive, hippocampal type of learning. We report that novel object location (nOL) learned at postnatal day 17 (PN17) undergoes the typical rapid forgetting of infantile learning. However, a later reminder reinstates memory expression. Furthermore, as for aversive experiences, nOL learning at PN17 engages critical period mechanisms in the dorsal hippocampus: it induces a switch in the GluN2A/2B-NMDA receptor ratio, and brain-derived neurotrophic factor injected bilaterally into the dorsal hippocampus immediately after training results in long-lasting memory expression. We conclude that in infancy the hippocampus plays a necessary role in processing episodic and contextual memories, including nonaversive ones, and matures through a developmental critical period.
Collapse
Affiliation(s)
- Alessio Travaglia
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Adam B Steinmetz
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Janelle M Miranda
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Cristina M Alberini
- Center for Neural Science, New York University, New York, New York 10003, USA
- Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016, USA
| |
Collapse
|
68
|
Janetsian-Fritz SS, Timme NM, Timm MM, McCane AM, Baucum Ii AJ, O'Donnell BF, Lapish CC. Maternal deprivation induces alterations in cognitive and cortical function in adulthood. Transl Psychiatry 2018; 8:71. [PMID: 29581432 PMCID: PMC5913289 DOI: 10.1038/s41398-018-0119-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/24/2017] [Accepted: 01/08/2018] [Indexed: 11/09/2022] Open
Abstract
Early life trauma is a risk factor for a number of neuropsychiatric disorders, including schizophrenia (SZ). The current study assessed how an early life traumatic event, maternal deprivation (MD), alters cognition and brain function in rodents. Rats were maternally deprived in the early postnatal period and then recognition memory (RM) was tested in adulthood using the novel object recognition task. The expression of catechol-o-methyl transferase (COMT) and glutamic acid decarboxylase (GAD67) were quantified in the medial prefrontal cortex (mPFC), ventral striatum, and temporal cortex (TC). In addition, depth EEG recordings were obtained from the mPFC, vertex, and TC during a paired-click paradigm to assess the effects of MD on sensory gating. MD animals exhibited impaired RM, lower expression of COMT in the mPFC and TC, and lower expression of GAD67 in the TC. Increased bioelectric noise was observed at each recording site of MD animals. MD animals also exhibited altered information theoretic measures of stimulus encoding. These data indicate that a neurodevelopmental perturbation yields persistent alterations in cognition and brain function, and are consistent with human studies that identified relationships between allelic differences in COMT and GAD67 and bioelectric noise. These changes evoked by MD also lead to alterations in shared information between cognitive and primary sensory processing areas, which provides insight into how early life trauma confers a risk for neurodevelopmental disorders, such as SZ, later in life.
Collapse
Affiliation(s)
- Sarine S Janetsian-Fritz
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA.
| | - Nicholas M Timme
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Maureen M Timm
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Aqilah M McCane
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Anthony J Baucum Ii
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Brian F O'Donnell
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Christopher C Lapish
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
- Indiana University School of Medicine Stark Neuroscience Institute, Indianapolis, IN, USA
- Indiana University-Purdue University Indianapolis School of Science Institute for Mathematical Modeling and Computational Sciences, Indianapolis, IN, USA
| |
Collapse
|
69
|
Sex differences in the effects of pre- and postnatal caffeine exposure on behavior and synaptic proteins in pubescent rats. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:416-425. [PMID: 28826637 DOI: 10.1016/j.pnpbp.2017.08.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 01/14/2023]
Abstract
Few studies have addressed the effects of caffeine in the puberty and/or adolescence in a sex dependent manner. Considering that caffeine intake has increased in this population, we investigated the behavioral and synaptic proteins changes in pubescent male and female rats after maternal consumption of caffeine. Adult female Wistar rats started to receive water or caffeine (0.1 and 0.3g/L in drinking water; low and moderate dose, respectively) during the active cycle at weekdays, two weeks before mating. The treatment lasted up to weaning and the offspring received caffeine until the onset of puberty (30-34days old). Behavioral tasks were performed to evaluate locomotor activity (open field task), anxious-like behavior (elevated plus maze task) and recognition memory (object recognition task) and synaptic proteins levels (proBDNF, BDNF, GFAP and SNAP-25) were verified in the hippocampus and cerebral cortex. While hyperlocomotion was observed in both sexes after caffeine treatment, anxiety-related behavior was attenuated by caffeine (0.3g/L) only in females. While moderate caffeine worsened recognition memory in females, an improvement in the long-term memory was observed in male rats for both doses. Coincident with memory improvement in males, caffeine increased pro- and BDNF in the hippocampus and cortex. Females presented increased proBDNF levels in both brain regions, with no effects of caffeine. While GFAP was not altered, moderate caffeine intake increased SNAP-25 in the cortex of female rats. Our findings revealed that caffeine promoted cognitive benefits in males associated with increased BDNF levels, while females showed less anxiety. Our findings revealed that caffeine promotes distinct behavioral outcomes and alterations in synaptic proteins during brain development in a sex dependent manner.
Collapse
|
70
|
Batista TH, Giusti-Paiva A, Vilela FC. Maternal protein malnutrition induces autism-like symptoms in rat offspring. Nutr Neurosci 2018; 22:655-663. [DOI: 10.1080/1028415x.2018.1427660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Tatiane Helena Batista
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Alexandre Giusti-Paiva
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Fabiana Cardoso Vilela
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| |
Collapse
|
71
|
Narducci R, Baroncelli L, Sansevero G, Begenisic T, Prontera C, Sale A, Cenni MC, Berardi N, Maffei L. Early impoverished environment delays the maturation of cerebral cortex. Sci Rep 2018; 8:1187. [PMID: 29352131 PMCID: PMC5775315 DOI: 10.1038/s41598-018-19459-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 12/27/2017] [Indexed: 12/26/2022] Open
Abstract
The influence of exposure to impoverished environments on brain development is unexplored since most studies investigated how environmental impoverishment affects adult brain. To shed light on the impact of early impoverishment on developmental trajectories of the nervous system, we developed a protocol of environmental impoverishment in which dams and pups lived from birth in a condition of reduced sensory-motor stimulation. Focusing on visual system, we measured two indexes of functional development, that is visual acuity, assessed by using Visual Evoked Potentials (VEPs), and VEP latency. In addition, we assessed in the visual cortex levels of Insulin-Like Growth Factor 1 (IGF-1) and myelin maturation, together with the expression of the GABA biosynthetic enzyme GAD67. We found that early impoverishment strongly delays visual acuity and VEP latency development. These functional changes were accompanied by a significant reduction of IGF-1 protein and GAD67 expression, as well as by delayed myelination of nerve fibers, in the visual cortex of impoverished pups. Thus, exposure to impoverished living conditions causes a significant alteration of developmental trajectories leading to a prominent delay of brain maturation. These results underscore the significance of adequate levels of environmental stimulation for the maturation of central nervous system.
Collapse
Affiliation(s)
- Roberta Narducci
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Area San Salvi - Pad. 26, I-50135, Florence, Italy
| | - Laura Baroncelli
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.
| | - Gabriele Sansevero
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Area San Salvi - Pad. 26, I-50135, Florence, Italy
| | - Tatjana Begenisic
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| | - Concetta Prontera
- Fondazione G. Monasterio CNR-Regione Toscana, via Moruzzi 1, I-56124, Pisa, Italy
| | - Alessandro Sale
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| | - Maria Cristina Cenni
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Area San Salvi - Pad. 26, I-50135, Florence, Italy
| | - Lamberto Maffei
- Institute of Neuroscience, National Research Council (CNR), Via Moruzzi 1, I-56124, Pisa, Italy
| |
Collapse
|
72
|
The α2C-adrenoceptor antagonist, ORM-10921, exerts antidepressant-like effects in the Flinders Sensitive Line rat. Behav Pharmacol 2018; 28:9-18. [PMID: 27749317 DOI: 10.1097/fbp.0000000000000261] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Depression involves deficits in monoaminergic neurotransmission. Differential roles for α2A, B and C subtypes of the α2-adrenoceptor (AR) are evident, with selective α2C-AR antagonists purported to have antidepressant and procognitive properties. However, this has not been demonstrated in a genetic animal model of depression. The role of the α2C-AR in modulating two key depression-related behaviours in the Flinders Sensitive Line (FSL) rat was studied using a dose-response analysis following subcutaneous administration with the selective α2C-AR antagonist ORM-10921 (0.03; 0.3 mg/kg), the nonselective α2-AR antagonist idazoxan (3 mg/kg), or vehicle once daily for 14 days. Behaviour in the novel object recognition test, forced swim test (FST) and locomotor activity test was assessed. To ratify the validity of the FSL model, the reference tricyclic antidepressant imipramine (15 mg/kg, intraperitoneally) was used as a comparator drug in the FST. FSL rats demonstrated significantly increased immobility and recognition memory deficits versus Flinders Resistant Line controls, with imipramine significantly reversing said immobility. Similarly, ORM-10921 at both doses but not idazoxan significantly reversed immobility in the FST as well as attenuated cognitive deficits in FSL animals. We conclude that selective α2C-AR antagonism has potential as a novel therapeutic strategy in the treatment of depression and cognitive dysfunction.
Collapse
|
73
|
Intra-nasal dopamine alleviates cognitive deficits in tgDISC1 rats which overexpress the human DISC1 gene. Neurobiol Learn Mem 2017; 146:12-20. [PMID: 29107702 DOI: 10.1016/j.nlm.2017.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 01/15/2023]
Abstract
The Disrupted-in-Schizophrenia 1 (DISC1) gene has been associated with mental illnesses such as major depression and schizophrenia. The transgenic DISC1 (tgDISC1) rat, which overexpresses the human DISC1 gene, is known to exhibit deficient dopamine (DA) homeostasis. To ascertain whether the DISC1 gene also impacts cognitive functions, 14-15 months old male tgDISC1 rats and wild-type controls were subjected to the novel object preference (NOP) test and the object-based attention test (OBAT) in order to assess short-term memory (1 h), long-term memory (24 h), and attention. RESULTS The tgDISC1 group exhibited intact short-term memory, but deficient long-term-memory in the NOP test and deficient attention-related behavior in the OBAT. In a different group of tgDISC1 rats, 3 mg/kg intranasally applied dopamine (IN-DA) or its vehicle was applied prior to the NOP or the OBAT test. IN-DA reversed cognitive deficits in both the NOP and OBAT tests. In a further cohort of tgDISC1 rats, post-mortem levels of DA, noradrenaline, serotonin and acetylcholine were determined in a variety of brain regions. The tgDISC1 group had less DA in the neostriatum, hippocampus and amygdala, less acetylcholine in neostriatum, nucleus accumbens, hippocampus, and amygdala, more serotonin in the nucleus accumbens, and less serotonin and noradrenaline in the amygdala. CONCLUSIONS Our findings show that DISC1 overexpression and misassembly is associated with deficits in long-term memory and attention-related behavior. Since behavioral impairments in tgDISC1 rats were reversed by IN-DA, DA deficiency may be a major cause for the behavioral deficits expressed in this model.
Collapse
|
74
|
Alterations in Mesoscopic Oscillations affecting Episodic Memory following Developmental Traumatic Brain Injury. Exp Neurol 2017; 300:259-273. [PMID: 29066322 DOI: 10.1016/j.expneurol.2017.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/27/2017] [Accepted: 10/20/2017] [Indexed: 11/20/2022]
Abstract
Certain deficits of episodic memory among young adults are the delayed consequences of an earlier mild or moderate Traumatic Brain Injury (mTBI). We examined alterations in hippocampal Local Field Oscillations (LFOs) of equivalently-impaired juvenile rodents to identify reliable functional markers of single-incidence mTBI. Two persistent, behavior-dependent, electrophysiological markers of injury were identified in the absence of external physiological symptoms by the analysis of wirelessly-transmitted hippocampal LFOs (3-80Hz) during repeat measures of the Novel Object Recognition (NOR) paradigm. Using a new method for detecting functional network activity at a single recording site, we correlated instantaneous increases in theta frequency and gamma magnitude with injury during periods of functional network organization and dissolution, presented here as Theta Epochs (TEs) and Theta Non-Epochs (TNEs), respectively. We estimated the efficacy of networks across pairs of such sites using new metrics such as the Coherence of Theta Phase (PCOH) and Inter-Epoch Intervals (IEIs) and demonstrated that behavioral deficits observed during the NOR testing stage correspond to electrophysiological deficits recorded during the preceding NOR familiarization stage. Increased theta frequency during TNEs and increased smoothness of PCOH during TEs were found to be robust markers of injury during memory-reliant behaviors.
Collapse
|
75
|
Cucarián JD, León LA, Luna GA, Torres MR, Corredor K, Cardenas P. F. CARACTERIZACIÓN TEMPORO-ESPACIAL DEL PATRÓN DE MARCHA EN ROEDORES COMO MODELO ANIMAL DE LESIÓN CEREBRAL CEREBROVASCULAR. ACTA BIOLÓGICA COLOMBIANA 2017. [DOI: 10.15446/abc.v22n3.65244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
En la investigación sobre movimiento, la experimentación animal ha proporcionado fundamentación científica para la investigación clínica, mejorando procedimientos diagnósticos y de rehabilitación. Lesiones cerebrales en roedores pueden ser usadas para modelar síntomas locomotores, sensoriales y/o cognitivos. Con el propósito de determinar la funcionalidad locomotriz y sensorial en roedores, se han propuesto varios métodos de evaluación y pronóstico clínico para identificar y evaluar adaptaciones estructurales y mecanismos de neuro-recuperación. Esto ha permitido que métodos de intervención terapéutica, como el ejercicio físico, sean utilizados para restaurar funciones sensitivo-motoras y cognitivas en roedores y humanos. La extrapolación (translación) de los resultados de investigaciones en ciencias básicas a áreas clínicas supone la continua cooperación y retroalimentación entre investigadores y profesionales de la salud, favoreciendo la formulación de intervenciones terapéuticas más eficaces basadas en resultados obtenidos de la experimentación animal. El objetivo de esta revisión es exponer las principales deficiencias motoras y los métodos empleados para determinar la dificultad motriz en la marcha en roedores con lesión cerebrovascular, para lo cual se realizó una revisión de literatura, sobre términos definidos (MeSH), en las bases de datos PsychINFO, Medline y Web of Science, entre enero de 2000 y enero de 2017. Se excluyeron artículos de carácter cualitativo o narrativo, sin revisión por pares, disertaciones, tesis o trabajos de grado y resúmenes de conferencias. Se revisan algunas manifestaciones clínicas, su efecto en la locomotricidad en roedores, algunas metodologías usadas para generar lesiones y para estudiar la función motriz, los principales métodos de medición y algunos aspectos translacionales.
Collapse
|
76
|
Gallant S, Welch L, Martone P, Shalev U. Effects of chronic prenatal MK-801 treatment on object recognition, cognitive flexibility, and drug-induced locomotor activity in juvenile and adult rat offspring. Behav Brain Res 2017; 328:62-69. [DOI: 10.1016/j.bbr.2017.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 03/31/2017] [Accepted: 04/01/2017] [Indexed: 12/29/2022]
|
77
|
Sta Maria NS, Reger ML, Cai Y, Baquing MAT, Buen F, Ponnaluri A, Hovda DA, Harris NG, Giza CC. D-Cycloserine Restores Experience-Dependent Neuroplasticity after Traumatic Brain Injury in the Developing Rat Brain. J Neurotrauma 2017; 34:1692-1702. [PMID: 27931146 PMCID: PMC5397224 DOI: 10.1089/neu.2016.4747] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) in children can cause persisting cognitive and behavioral dysfunction, and inevitably raises concerns about lost potential in these injured youth. Lateral fluid percussion injury (FPI) in weanling rats pathologically affects hippocampal N-methyl-d-aspartate receptor (NMDAR)- and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated glutamatergic neurotransmission subacutely within the first post-injury week. FPI to weanling rats has also been shown to impair enriched-environment (EE) induced enhancement of Morris water maze (MWM) learning and memory in adulthood. Recently, improved outcomes can be achieved using agents that enhance NMDAR function. We hypothesized that administering D-cycloserine (DCS), an NMDAR co-agonist, every 12 h (i.p.) would restore subacute glutamatergic neurotransmission and reinstate experience-dependent plasticity. Postnatal day 19 (P19) rats received either a sham or FPI. On post-injury day (PID) 1-3, animals were randomized to saline (Sal) or DCS. Firstly, immunoblotting of hippocampal NMDAR and AMPAR proteins were measured on PID4. Second, PID4 novel object recognition, an NMDAR- and hippocampal- mediated working memory task, was assessed. Third, P19 rats were placed in an EE (17 days), and MWM performance was measured, starting on PID30. On PID4, DCS restored reduced NR2A and increased GluR2 by 54%, and also restored diminished recognition memory in FPI pups. EE significantly improved MWM performance in shams, regardless of treatment. In contrast, FPI-EE-Sal animals only performed to the level of standard housed animals, whereas FPI-EE-DCS animals were comparable with sham-EE counterparts. This study shows that NMDAR agonist use during reduced glutamatergic transmission after developmental TBI can reinstate early molecular and behavioral responses that subsequently manifest in experience-dependent plasticity and rescued potential.
Collapse
Affiliation(s)
- Naomi S. Sta Maria
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
- Department of Bioengineering, UCLA Brain Injury Research Center, Los Angeles, California
| | - Maxine L. Reger
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
- Department of Psychology, UCLA Brain Injury Research Center, Los Angeles, California
| | - Yan Cai
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
| | - Mary Anne T. Baquing
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
- Harbor-UCLA Department of Obstetrics and Gynecology, UCLA Brain Injury Research Center, Los Angeles, California
| | - Floyd Buen
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
- Department of Head and Neck Surgery, UCLA Brain Injury Research Center, Los Angeles, California
| | - Aditya Ponnaluri
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
- Department of Mechanical Engineering, UCLA Brain Injury Research Center, Los Angeles, California
| | - David A. Hovda
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
- Department of Medical and Molecular Pharmacology, UCLA Brain Injury Research Center, Los Angeles, California
| | - Neil G. Harris
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
| | - Christopher C. Giza
- Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, California
- Division of Pediatric Neurology, UCLA Brain Injury Research Center, Los Angeles, California
| |
Collapse
|
78
|
Duque A, Vinader-Caerols C, Monleón S. Indomethacin counteracts the effects of chronic social defeat stress on emotional but not recognition memory in mice. PLoS One 2017; 12:e0173182. [PMID: 28278165 PMCID: PMC5344348 DOI: 10.1371/journal.pone.0173182] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/16/2017] [Indexed: 12/15/2022] Open
Abstract
We have previously observed the impairing effects of chronic social defeat stress (CSDS) on emotional memory in mice. Given the relation between stress and inflammatory processes, we sought to study the effectiveness of the anti-inflammatory indomethacin in reversing the detrimental effects of CSDS on emotional memory in mice. The effects of CSDS and indomethacin on recognition memory were also evaluated. Male CD1 mice were randomly divided into four groups: non-stressed + saline (NS+SAL); non-stressed + indomethacin (NS+IND); stressed + saline (S+SAL); and stressed + indomethacin (S+IND). Stressed animals were exposed to a daily 10 min agonistic confrontation (CSDS) for 20 days. All subjects were treated daily with saline or indomethacin (10 mg/kg, i.p.). 24 h after the CSDS period, all the mice were evaluated in a social interaction test to distinguish between those that were resilient or susceptible to social stress. All subjects (n = 10–12 per group) were then evaluated in inhibitory avoidance (IA), novel object recognition (NOR), elevated plus maze and hot plate tests. As in control animals (NS+SAL group), IA learning was observed in the resilient groups, as well as in the susceptible mice treated with indomethacin (S+IND group). Recognition memory was observed in the non-stressed and the resilient mice, but not in the susceptible animals. Also, stressed mice exhibited higher anxiety levels. No significant differences were observed in locomotor activity or analgesia. In conclusion, CSDS induces anxiety in post-pubertal mice and impairs emotional and recognition memory in the susceptible subjects. The effects of CSDS on emotional memory, but not on recognition memory and anxiety, are reversed by indomethacin. Moreover, memory impairment is not secondary to the effects of CSDS on locomotor activity, emotionality or pain sensitivity.
Collapse
Affiliation(s)
- Aránzazu Duque
- Department of Psychobiology, University of Valencia, Valencia, Spain
| | | | - Santiago Monleón
- Department of Psychobiology, University of Valencia, Valencia, Spain
- * E-mail:
| |
Collapse
|
79
|
Melancia F, Servadio M, Schiavi S, Campolongo P, Giusti-Paiva A, Trezza V. Testing the correlation between experimentally-induced hypothyroidism during pregnancy and autistic-like symptoms in the rat offspring. Behav Brain Res 2017; 321:113-122. [DOI: 10.1016/j.bbr.2016.12.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 12/19/2016] [Accepted: 12/23/2016] [Indexed: 12/17/2022]
|
80
|
Tang S, Xu S, Lu X, Gullapalli RP, McKenna MC, Waddell J. Neuroprotective Effects of Acetyl-L-Carnitine on Neonatal Hypoxia Ischemia-Induced Brain Injury in Rats. Dev Neurosci 2017; 38:384-396. [PMID: 28226317 DOI: 10.1159/000455041] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/12/2016] [Indexed: 12/17/2022] Open
Abstract
Perinatal hypoxia ischemia (HI) is a significant cause of brain injury in surviving infants. Although hypothermia improves outcomes in some infants, additional therapies are needed since about 40% of infants still have a poor outcome. Acetyl-L-carnitine (ALCAR), an acetylated derivative of L-carnitine, protected against early changes in brain metabolites and mitochondrial function after HI on postnatal day (PND) 7 in a rat pup model of near-term HI injury. However, its efficacy in long-term structural and functional outcomes remains unexplored. We determined the efficacy of ALCAR therapy administered to rat pups after HI at PND 7, using both longitudinal in vivo magnetic resonance imaging and behavioral tests, in male and female rats. HI led to sex-specific behavioral impairment, with males exhibiting more global functional deficits than females. Interestingly, HI reduced the volume of the contralateral hemisphere in males only, suggesting that the brain injury is more diffuse in males than in females. Treatment with ALCAR improved both morphological and functional outcomes in both male and female rats. These results suggest that ALCAR may be a potential therapy for clinical use since the treatment attenuated the moderate injury produced under the experimental conditions used and improved the functional outcome in preclinical studies.
Collapse
Affiliation(s)
- Shiyu Tang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | |
Collapse
|
81
|
Neuroprotective Effects of Aged Garlic Extract on Cognitive Dysfunction and Neuroinflammation Induced by β-Amyloid in Rats. Nutrients 2017; 9:nu9010024. [PMID: 28054940 PMCID: PMC5295068 DOI: 10.3390/nu9010024] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/05/2016] [Accepted: 12/26/2016] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is pathological evidence of Alzheimer's disease (AD) that likely starts as a host defense response to the damaging effects of the β-amyloid (Aβ) deposits in the brain. The activation of microglia may promote the neurodegenerative process through the release of proinflammatory cytokines, such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNFα), which may lead to neuronal damage and eventual death. Aged garlic extract (AGE) has been reported to have multiple biological activities, including anti-inflammatory effects. Therefore, the objective of this study was to investigate the effect of AGE on Aβ (1-42)-induced cognitive dysfunction and neuroinflammation. Adult male Wistar rats were given AGE (125, 250, and 500 mg/kg BW, body weight), orally administered, daily for 56 days. They were then injected with 1 μL of aggregated Aβ (1-42) into the lateral ventricles; bilaterally. Seven days later, their recognition memory was evaluated using a novel object recognition (NOR) test. Then the rats were sacrificed to investigate the alteration of microglia cells, IL-1β and TNFα in the cerebral cortex and hippocampus. The results indicated that AGE at doses of 250 and 500 mg/kg BW significantly improved short-term recognition memory in cognitively impaired rats. In addition, AGE significantly minimized the inflammatory response by reducing the activation of microglia and IL-1β to the levels found in the control, which is similar to the results found in Celebrex-treated rats. In conclusion, AGE may be useful for improving the short-term recognition memory and relieve the neuroinflammation in Aβ-induced rats.
Collapse
|
82
|
Uys MM, Shahid M, Harvey BH. Therapeutic Potential of Selectively Targeting the α 2C-Adrenoceptor in Cognition, Depression, and Schizophrenia-New Developments and Future Perspective. Front Psychiatry 2017; 8:144. [PMID: 28855875 PMCID: PMC5558054 DOI: 10.3389/fpsyt.2017.00144] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 07/24/2017] [Indexed: 12/12/2022] Open
Abstract
α2A- and α2C-adrenoceptors (ARs) are the primary α2-AR subtypes involved in central nervous system (CNS) function. These receptors are implicated in the pathophysiology of psychiatric illness, particularly those associated with affective, psychotic, and cognitive symptoms. Indeed, non-selective α2-AR blockade is proposed to contribute toward antidepressant (e.g., mirtazapine) and atypical antipsychotic (e.g., clozapine) drug action. Both α2C- and α2A-AR share autoreceptor functions to exert negative feedback control on noradrenaline (NA) release, with α2C-AR heteroreceptors regulating non-noradrenergic transmission (e.g., serotonin, dopamine). While the α2A-AR is widely distributed throughout the CNS, α2C-AR expression is more restricted, suggesting the possibility of significant differences in how these two receptor subtypes modulate regional neurotransmission. However, the α2C-AR plays a more prominent role during states of low endogenous NA activity, while the α2A-AR is relatively more engaged during states of high noradrenergic tone. Although augmentation of conventional antidepressant and antipsychotic therapy with non-selective α2-AR antagonists may improve therapeutic outcome, animal studies report distinct yet often opposing roles for the α2A- and α2C-ARs on behavioral markers of mood and cognition, implying that non-selective α2-AR antagonism may compromise therapeutic utility both in terms of efficacy and side-effect liability. Recently, several highly selective α2C-AR antagonists have been identified that have allowed deeper investigation into the function and utility of the α2C-AR. ORM-13070 is a useful positron emission tomography ligand, ORM-10921 has demonstrated antipsychotic, antidepressant, and pro-cognitive actions in animals, while ORM-12741 is in clinical development for the treatment of cognitive dysfunction and neuropsychiatric symptoms in Alzheimer's disease. This review will emphasize the importance and relevance of the α2C-AR as a neuropsychiatric drug target in major depression, schizophrenia, and associated cognitive deficits. In addition, we will present new prospects and future directions of investigation.
Collapse
Affiliation(s)
- Madeleine Monique Uys
- Division of Pharmacology, Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | | | - Brian Herbert Harvey
- Division of Pharmacology, Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
83
|
Goepfrich AA, Friemel CM, Pauen S, Schneider M. Ontogeny of sensorimotor gating and short-term memory processing throughout the adolescent period in rats. Dev Cogn Neurosci 2016; 25:167-175. [PMID: 27908562 PMCID: PMC6987840 DOI: 10.1016/j.dcn.2016.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 11/02/2016] [Accepted: 11/15/2016] [Indexed: 11/17/2022] Open
Abstract
Adolescence and puberty are highly susceptible developmental periods during which the neuronal organization and maturation of the brain is completed. The endocannabinoid (eCB) system, which is well known to modulate cognitive processing, undergoes profound and transient developmental changes during adolescence. With the present study we were aiming to examine the ontogeny of cognitive skills throughout adolescence in male rats and clarify the potential modulatory role of CB1 receptor signalling. Cognitive skills were assessed repeatedly every 10th day in rats throughout adolescence. All animals were tested for object recognition memory and prepulse inhibition of the acoustic startle reflex. Although cognitive performance in short-term memory as well as sensorimotor gating abilities were decreased during puberty compared to adulthood, both tasks were found to show different developmental trajectories throughout adolescence. A low dose of the CB1 receptor antagonist/inverse agonist SR141716 was found to improve recognition memory specifically in pubertal animals while not affecting behavioral performance at other ages tested. The present findings demonstrate that the developmental trajectory of cognitive abilities does not occur linearly for all cognitive processes and is strongly influenced by pubertal maturation. Developmental alterations within the eCB system at puberty onset may be involved in these changes in cognitive processing.
Collapse
Affiliation(s)
- Anja A Goepfrich
- Research Group Developmental Neuropsychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Chris M Friemel
- Research Group Developmental Neuropsychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sabina Pauen
- Department of Psychology, University of Heidelberg, Germany
| | | |
Collapse
|
84
|
Ramsaran AI, Sanders HR, Stanton ME. Determinants of object-in-context and object-place-context recognition in the developing rat. Dev Psychobiol 2016; 58:883-895. [DOI: 10.1002/dev.21432] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/10/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Adam I. Ramsaran
- Department of Psychological and Brain Sciences; University of Delaware; Newark Delaware
| | - Hollie R. Sanders
- Department of Psychological and Brain Sciences; University of Delaware; Newark Delaware
| | - Mark E. Stanton
- Department of Psychological and Brain Sciences; University of Delaware; Newark Delaware
| |
Collapse
|
85
|
Bailey ZS, Grinter MB, VandeVord PJ. Astrocyte Reactivity Following Blast Exposure Involves Aberrant Histone Acetylation. Front Mol Neurosci 2016; 9:64. [PMID: 27551260 PMCID: PMC4976110 DOI: 10.3389/fnmol.2016.00064] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/21/2016] [Indexed: 12/17/2022] Open
Abstract
Blast induced neurotrauma (BINT) is a prevalent injury within military and civilian populations. The injury is characterized by persistent inflammation at the cellular level which manifests as a multitude of cognitive and functional impairments. Epigenetic regulation of transcription offers an important control mechanism for gene expression and cellular function which may underlie chronic inflammation and result in neurodegeneration. We hypothesize that altered histone acetylation patterns may be involved in blast induced inflammation and the chronic activation of glial cells. This study aimed to elucidate changes to histone acetylation occurring following injury and the roles these changes may have within the pathology. Sprague Dawley rats were subjected to either a 10 or 17 psi blast overpressure within an Advanced Blast Simulator (ABS). Sham animals underwent the same procedures without blast exposure. Memory impairments were measured using the Novel Object Recognition (NOR) test at 2 and 7 days post-injury. Tissues were collected at 7 days for Western blot and immunohistochemistry (IHC) analysis. Sham animals showed intact memory at each time point. The novel object discrimination decreased significantly between two and 7 days for each injury group (p < 0.05). This is indicative of the onset of memory impairment. Western blot analysis showed glial fibrillary acidic protein (GFAP), a known marker of activated astrocytes, was elevated in the prefrontal cortex (PFC) following blast exposure for both injury groups. Analysis of histone protein extract showed no changes in the level of any total histone proteins within the PFC. However, acetylation levels of histone H2b, H3, and H4 were decreased in both groups (p < 0.05). Co-localization immunofluorescence was used to further investigate any potential correlation between decreased histone acetylation and astrocyte activation. These experiments showed a similar decrease in H3 acetylation in astrocytes exposed to a 17 psi blast but not a 10 psi blast. Further investigation of gene expression by polymerase chain reaction (PCR) array, showed dysregulation of several cytokine and cytokine receptors that are involved in neuroinflammatory processes. We have shown aberrant histone acetylation patterns involved in blast induced astrogliosis and cognitive impairments. Further understanding of their role in the injury progression may lead to novel therapeutic targets.
Collapse
Affiliation(s)
- Zachary S Bailey
- Department of Biomedical Engineering and Mechanics, Virginia Tech Blacksburg, VA, USA
| | - Michael B Grinter
- Department of Biomedical Engineering and Mechanics, Virginia Tech Blacksburg, VA, USA
| | - Pamela J VandeVord
- Department of Biomedical Engineering and Mechanics, Virginia TechBlacksburg, VA, USA; Salem Veterans Affairs Medical CenterSalem, VA, USA
| |
Collapse
|
86
|
Perinatal reduction of functional serotonin transporters results in developmental delay. Neuropharmacology 2016; 109:96-111. [PMID: 27208789 DOI: 10.1016/j.neuropharm.2016.05.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 03/25/2016] [Accepted: 05/17/2016] [Indexed: 11/22/2022]
Abstract
While there is strong evidence from rodent and human studies that a reduction in serotonin transporter (5-HTT) function in early-life can increase the risk for several neuropsychiatric disorders in adulthood, the effects of reduced 5-HTT function on behavior across developmental stages are underinvestigated. To elucidate how perinatal pharmacological and lifelong genetic inactivation of the 5-HTT affects behavior across development, we conducted a battery of behavioral tests in rats perinatally exposed to fluoxetine or vehicle and in 5-HTT(-/-) versus 5-HTT(+/+) rats. We measured motor-related behavior, olfactory function, grooming behavior, sensorimotor gating, object directed behavior and novel object recognition in the first three postnatal weeks and if possible the tests were repeated in adolescence and adulthood. We also measured developmental milestones such as eye opening, reflex development and body weight. We observed that both pharmacological and genetic inactivation of 5-HTT resulted in a developmental delay. Except for hypo-locomotion, most of the observed early-life effects were normalized later in life. In adolescence and adulthood we observed object directed behavior and decreased novel object recognition in the 5-HTT(-/-) rats, which might be related to the lifelong inactivation of 5-HTT. Together, these data provide an important contribution to the understanding of the effects of perinatal and lifelong 5-HTT inactivation on behavior across developmental stages.
Collapse
|
87
|
Schober ME, Requena DF, Abdullah OM, Casper TC, Beachy J, Malleske D, Pauly JR. Dietary Docosahexaenoic Acid Improves Cognitive Function, Tissue Sparing, and Magnetic Resonance Imaging Indices of Edema and White Matter Injury in the Immature Rat after Traumatic Brain Injury. J Neurotrauma 2016; 33:390-402. [PMID: 26247583 PMCID: PMC4761828 DOI: 10.1089/neu.2015.3945] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of acquired neurologic disability in children. Specific therapies to treat acute TBI are lacking. Cognitive impairment from TBI may be blunted by decreasing inflammation and oxidative damage after injury. Docosahexaenoic acid (DHA) decreases cognitive impairment, oxidative stress, and white matter injury in adult rats after TBI. Effects of DHA on cognitive outcome, oxidative stress, and white matter injury in the developing rat after experimental TBI are unknown. We hypothesized that DHA would decrease early inflammatory markers and oxidative stress, and improve cognitive, imaging and histologic outcomes in rat pups after controlled cortical impact (CCI). CCI or sham surgery was delivered to 17 d old male rat pups exposed to DHA or standard diet for the duration of the experiments. DHA was introduced into the dam diet the day before CCI to allow timely DHA delivery to the pre-weanling pups. Inflammatory cytokines and nitrates/nitrites were measured in the injured brains at post-injury Day (PID) 1 and PID2. Morris water maze (MWM) testing was performed at PID41-PID47. T2-weighted and diffusion tensor imaging studies were obtained at PID12 and PID28. Tissue sparing was calculated histologically at PID3 and PID50. DHA did not adversely affect rat survival or weight gain. DHA acutely decreased oxidative stress and increased anti-inflammatory interleukin 10 in CCI brains. DHA improved MWM performance and lesion volume late after injury. At PID12, DHA decreased T2-imaging measures of cerebral edema and decreased radial diffusivity, an index of white matter injury. DHA improved short- and long-term neurologic outcomes after CCI in the rat pup. Given its favorable safety profile, DHA is a promising candidate therapy for pediatric TBI. Further studies are needed to explore neuroprotective mechanisms of DHA after developmental TBI.
Collapse
Affiliation(s)
- Michelle E Schober
- 1 Department of Pediatrics, Division of Critical Care, University of Utah , Salt Lake City, Utah
| | - Daniela F Requena
- 1 Department of Pediatrics, Division of Critical Care, University of Utah , Salt Lake City, Utah
| | - Osama M Abdullah
- 2 Department of Bioengineering, University of Utah , Salt Lake City, Utah
| | - T Charles Casper
- 1 Department of Pediatrics, Division of Critical Care, University of Utah , Salt Lake City, Utah
| | - Joanna Beachy
- 3 Department of Pediatrics, Division of Neonatology, University of Utah , Salt Lake City, Utah
| | - Daniel Malleske
- 3 Department of Pediatrics, Division of Neonatology, University of Utah , Salt Lake City, Utah
| | - James R Pauly
- 4 College of Pharmacy and Spinal Cord and Brain Injury Research Center, University of Kentucky , Lexington, Kentucky
| |
Collapse
|
88
|
Smith CJW, Wilkins KB, Mogavero JN, Veenema AH. Social Novelty Investigation in the Juvenile Rat: Modulation by the μ-Opioid System. J Neuroendocrinol 2015. [PMID: 26212131 DOI: 10.1111/jne.12301] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The drive to approach and explore novel conspecifics is inherent to social animals and may promote optimal social functioning. Juvenile animals seek out interactions with novel peers more frequently and find these interactions to be more rewarding than their adult counterparts. In the present study, we aimed to establish a behavioural paradigm to measure social novelty-seeking in juvenile rats and to determine the involvement of the opioid, dopamine, oxytocin and vasopressin systems in this behaviour. To this end, we developed the social novelty preference test to assess the preference of a juvenile rat to investigate a novel over a familiar (cage mate) conspecific. We show that across the juvenile period both male and female rats spend more time investigating a novel conspecific than a cage mate, independent of subject sex or repeated exposure to the test. We hypothesised that brain systems subserving social information processing and social motivation/reward (i.e. the opioid, dopamine, oxytocin, vasopressin systems) might support social novelty preference. To test this, receptor antagonists of each of these systems were administered i.c.v. prior to exposure to the social novelty preference test and, subsequently, to the social preference test, to examine the specificity of these effects. We find that μ-opioid receptor antagonism reduces novel social investigation in both the social novelty preference and social preference tests while leaving the investigation of a cage mate (social novelty preference test) or an object (social preference test) unaffected. In contrast, central blockade of dopamine D2 receptors (with eticlopride), oxytocin receptors (with des-Gly-NH2,d(CH2)5[Tyr(Me)2,Thr4]OVT) or vasopressin V1a receptors [with (CH2)5Tyr(Me2)AVP] failed to alter social novelty preference or social preference. Overall, we have established a new behavioural test to study social novelty-seeking behaviour in the juvenile rat and show that the μ-opioid system facilitates this behaviour, possibly by reducing risk avoidance and enhancing the hedonic and/or motivational value of social novelty.
Collapse
Affiliation(s)
- C J W Smith
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - K B Wilkins
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - J N Mogavero
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - A H Veenema
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| |
Collapse
|
89
|
May Z, Morrill A, Holcombe A, Johnston T, Gallup J, Fouad K, Schalomon M, Hamilton TJ. Object recognition memory in zebrafish. Behav Brain Res 2015; 296:199-210. [PMID: 26376244 DOI: 10.1016/j.bbr.2015.09.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/08/2015] [Accepted: 09/10/2015] [Indexed: 01/19/2023]
Abstract
The novel object recognition, or novel-object preference (NOP) test is employed to assess recognition memory in a variety of organisms. The subject is exposed to two identical objects, then after a delay, it is placed back in the original environment containing one of the original objects and a novel object. If the subject spends more time exploring one object, this can be interpreted as memory retention. To date, this test has not been fully explored in zebrafish (Danio rerio). Zebrafish possess recognition memory for simple 2- and 3-dimensional geometrical shapes, yet it is unknown if this translates to complex 3-dimensional objects. In this study we evaluated recognition memory in zebrafish using complex objects of different sizes. Contrary to rodents, zebrafish preferentially explored familiar over novel objects. Familiarity preference disappeared after delays of 5 mins. Leopard danios, another strain of D. rerio, also preferred the familiar object after a 1 min delay. Object preference could be re-established in zebra danios by administration of nicotine tartrate salt (50mg/L) prior to stimuli presentation, suggesting a memory-enhancing effect of nicotine. Additionally, exploration biases were present only when the objects were of intermediate size (2 × 5 cm). Our results demonstrate zebra and leopard danios have recognition memory, and that low nicotine doses can improve this memory type in zebra danios. However, exploration biases, from which memory is inferred, depend on object size. These findings suggest zebrafish ecology might influence object preference, as zebrafish neophobia could reflect natural anti-predatory behaviour.
Collapse
Affiliation(s)
- Zacnicte May
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada; Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, AB T6G 2G4, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Adam Morrill
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Adam Holcombe
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Travis Johnston
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Joshua Gallup
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Karim Fouad
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, AB T6G 2G4, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Melike Schalomon
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Trevor James Hamilton
- Department of Psychology, MacEwan University, Edmonton, AB T5J 4S2, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada,.
| |
Collapse
|
90
|
Sex differences in cell genesis, hippocampal volume and behavioral outcomes in a rat model of neonatal HI. Exp Neurol 2015; 275 Pt 2:285-95. [PMID: 26376217 DOI: 10.1016/j.expneurol.2015.09.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/27/2015] [Accepted: 09/06/2015] [Indexed: 12/11/2022]
Abstract
Hypoxia-ischemia (HI) of the brain in near-term and term infants is a leading cause of infant mortality and lifelong disability but current therapeutic approaches remain limited. Males consistently display greater vulnerability to the deleterious consequences of HI in both humans and animal models. Neurogenesis increases after neonatal HI and offers a potential therapeutic target for recovery. The steroid hormone estradiol has been extensively explored as a neuroprotectant in adult models of stroke but with mixed results. Less consideration has been afforded to this naturally occurring agent in the developing brain, which has unique challenges from the adult. Using a model of term HI in the rat we have explored the impact of this insult on cell genesis in the hippocampus of males and females and the ability of estradiol treatment immediately after insult to restore function. Both short-term (3 days) and long-term (7 days) post-injury were assessed and revealed that only females had markedly increased cell genesis on the short-term but both sexes were increased long-term. A battery of behavioral tests revealed motor impairment in males and compromised episodic memory while both sexes were modestly impaired in spatial memory. Juvenile social play was also depressed in both sexes after HI. Estradiol therapy improved behavioral performance in both sexes but did not reverse a deficit in hippocampal volume ipsilateral to the insult. Thus the effects of estradiol do not appear to be via cell death or proliferation but rather involve other components of neural functioning.
Collapse
|
91
|
Therapeutic hypothermia and hypoxia-ischemia in the term-equivalent neonatal rat: characterization of a translational preclinical model. Pediatr Res 2015; 78:264-71. [PMID: 25996893 PMCID: PMC4543535 DOI: 10.1038/pr.2015.100] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/24/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is a major cause of morbidity in survivors. Therapeutic hypothermia (TH) is the only available intervention, but the protection is incomplete. Preclinical studies of HIE/TH in the rodent have relied on the postnatal day (P) 7 rat whose brain approximates a 32-36 wk gestation infant, less relevant for these studies. We propose that HIE and TH in the term-equivalent P10 rat will be more translational. METHODS P10-11 rat pups were subjected to unilateral hypoxia-ischemia (HI) and 4 h recovery in normothermic (N) or hypothermic (TH) conditions. Brain damage was assessed longitudinally at 24 h, 2 wk, and 12 wk. Motor function was assessed with the beam walk; recognition memory was measured by novel object recognition. RESULTS Neuroprotection with TH was apparent at 2 and 12 wk in both moderately and severely damaged animals. TH improved motor function in moderate, but not severe, damage. Impaired object recognition occurred with severe damage with no evidence of protection of TH. CONCLUSION This adaptation of the immature rat model of HI provides a reproducible platform to further study HIE/TH in which individual animals are followed up longitudinally to provide a useful translational preclinical model.
Collapse
|
92
|
Mustafar F, De Luna P, Rainer G. Enhanced visual exploration for real objects compared to pictures during free viewing in the macaque monkey. Behav Processes 2015; 118:8-20. [DOI: 10.1016/j.beproc.2015.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/19/2015] [Accepted: 05/19/2015] [Indexed: 01/23/2023]
|
93
|
Zhao QR, Lu JM, Yao JJ, Zhang ZY, Ling C, Mei YA. Neuritin reverses deficits in murine novel object associative recognition memory caused by exposure to extremely low-frequency (50 Hz) electromagnetic fields. Sci Rep 2015; 5:11768. [PMID: 26138388 PMCID: PMC4650637 DOI: 10.1038/srep11768] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 06/01/2015] [Indexed: 12/21/2022] Open
Abstract
Animal studies have shown that electromagnetic field exposure may interfere with the activity of brain cells, thereby generating behavioral and cognitive disturbances. However, the underlying mechanisms and possible preventions are still unknown. In this study, we used a mouse model to examine the effects of exposure to extremely low-frequency (50 Hz) electromagnetic fields (ELF MFs) on a recognition memory task and morphological changes of hippocampal neurons. The data showed that ELF MFs exposure (1 mT, 12 h/day) induced a time-dependent deficit in novel object associative recognition memory and also decreased hippocampal dendritic spine density. This effect was observed without corresponding changes in spontaneous locomotor activity and was transient, which has only been seen after exposing mice to ELF MFs for 7-10 days. The over-expression of hippocampal neuritin, an activity-dependent neurotrophic factor, using an adeno-associated virus (AAV) vector significantly increased the neuritin level and dendritic spine density. This increase was paralleled with ELF MFs exposure-induced deficits in recognition memory and reductions of dendritic spine density. Collectively, our study provides evidence for the association between ELF MFs exposure, impairment of recognition memory, and resulting changes in hippocampal dendritic spine density. Neuritin prevented this ELF MFs-exposure-induced effect by increasing the hippocampal spine density.
Collapse
Affiliation(s)
- Qian-Ru Zhao
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Jun-Mei Lu
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Jin-Jing Yao
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Zheng-Yu Zhang
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Yan-Ai Mei
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| |
Collapse
|
94
|
Raber J. Novel images and novel locations of familiar images as sensitive translational cognitive tests in humans. Behav Brain Res 2015; 285:53-9. [DOI: 10.1016/j.bbr.2015.01.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 01/20/2023]
|
95
|
Ramsaran AI, Westbrook SR, Stanton ME. Ontogeny of object-in-context recognition in the rat. Behav Brain Res 2015; 298:37-47. [PMID: 25892362 DOI: 10.1016/j.bbr.2015.04.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/27/2015] [Accepted: 04/08/2015] [Indexed: 02/06/2023]
Abstract
The object-in-context recognition (OiC) task [19] is a spontaneous exploration task that serves as an index of incidental contextual learning and memory. During the test phase, rats prefer to explore the object mismatched to the testing context based on previous object-context pairings experienced during training. The mechanisms of OiC memory have been explored in adult rats [12,35]; however, little is known about its determinants during development. Thus, the present study examined the ontogeny of the OiC task in preweanling through adolescent rats. We demonstrate that postnatal day (PD) 17, 21, 26, and 31 rats can perform the OiC task (Experiment 1) and that preference for the novel target is eliminated when rats are tested in an alternate context not encountered during training (Experiment 2). Lastly, we show that PD26 but not PD17 rats can perform the OiC task when the training contexts only differed by distal spatial cues (Experiment 3). These data demonstrate for the first time that PD17 rats can acquire and retain short-term OiC memory, which involves associative learning of object and context information. However, we also provide evidence that preweanling rats' ability to utilize certain aspects of a context (i.e., distal spatial cues) in the OiC task is not equivalent to that of their older counterparts. Implications for the development of contextual memory and its related neural substrates are discussed.
Collapse
|
96
|
Schober ME, Requena DF, Block B, Davis LJ, Rodesch C, Casper TC, Juul SE, Kesner RP, Lane RH. Erythropoietin improved cognitive function and decreased hippocampal caspase activity in rat pups after traumatic brain injury. J Neurotrauma 2014; 31:358-69. [PMID: 23972011 DOI: 10.1089/neu.2013.2922] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Traumatic brain injury (TBI) is a leading cause of acquired neurologic disability in children. Erythropoietin (EPO), an anti-apoptotic cytokine, improved cognitive outcome in adult rats after TBI. To our knowledge, EPO has not been studied in a developmental TBI model. HYPOTHESIS We hypothesized that EPO would improve cognitive outcome and increase neuron fraction in the hippocampus in 17-day-old (P17) rat pups after controlled cortical impact (CCI). METHODS EPO or vehicle was given at 1, 24, and 48 h after CCI and at post injury day (PID) 7. Cognitive outcome at PID14 was assessed using Novel Object Recognition (NOR). Hippocampal EPO levels, caspase activity, and mRNA levels of the apoptosis factors Bcl2, Bax, Bcl-xL, and Bad were measured during the first 14 days after injury. Neuron fraction and caspase activation in CA1, CA3, and DG were studied at PID2. RESULTS EPO normalized recognition memory after CCI. EPO blunted the increased hippocampal caspase activity induced by CCI at PID1, but not at PID2. EPO increased neuron fraction in CA3 at PID2. Brain levels of exogenous EPO appeared low relative to endogenous. Timing of EPO administration was associated with temporal changes in hippocampal mRNA levels of EPO and pro-apoptotic factors. Conclusion/Speculation: EPO improved recognition memory, increased regional hippocampal neuron fraction, and decreased caspase activity in P17 rats after CCI. We speculate that EPO improved cognitive outcome in rat pups after CCI as a result of improved neuronal survival via inhibition of caspase-dependent apoptosis early after injury.
Collapse
Affiliation(s)
- Michelle E Schober
- 1 Department of Pediatrics, Division of Critical Care, University of Utah , Salt Lake City, Utah
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Westbrook SR, Brennan LE, Stanton ME. Ontogeny of object versus location recognition in the rat: acquisition and retention effects. Dev Psychobiol 2014; 56:1492-506. [PMID: 24992011 DOI: 10.1002/dev.21232] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 05/27/2014] [Indexed: 12/11/2022]
Abstract
Novel object and location recognition tasks harness the rat's natural tendency to explore novelty (Berlyne, 1950) to study incidental learning. The present study examined the ontogenetic profile of these two tasks and retention of spatial learning between postnatal day (PD) 17 and 31. Experiment 1 showed that rats ages PD17, 21, and 26 recognize novel objects, but only PD21 and PD26 rats recognize a novel location of a familiar object. These results suggest that novel object recognition develops before PD17, while object location recognition emerges between PD17 and PD21. Experiment 2 studied the ontogenetic profile of object location memory retention in PD21, 26, and 31 rats. PD26 and PD31 rats retained the object location memory for both 10-min and 24-hr delays. PD21 rats failed to retain the object location memory for the 24-hr delay, suggesting differential development of short- versus long-term memory in the ontogeny of object location memory.
Collapse
Affiliation(s)
- Sara R Westbrook
- Department of Psychology, University of Delaware, Newark, Delaware, 19716
| | | | | |
Collapse
|
98
|
Ardais A, Borges M, Rocha A, Sallaberry C, Cunha R, Porciúncula L. Caffeine triggers behavioral and neurochemical alterations in adolescent rats. Neuroscience 2014; 270:27-39. [DOI: 10.1016/j.neuroscience.2014.04.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 03/31/2014] [Accepted: 04/01/2014] [Indexed: 12/01/2022]
|
99
|
Cognitive impairment and dentate gyrus synaptic dysfunction in experimental parkinsonism. Biol Psychiatry 2014; 75:701-10. [PMID: 23541633 DOI: 10.1016/j.biopsych.2013.02.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/20/2013] [Accepted: 02/20/2013] [Indexed: 11/21/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by the progressive degeneration of the nigrostriatal dopaminergic pathway and the emergence of rigidity, tremor, and bradykinesia. Accumulating evidence indicates that PD is also accompanied by nonmotor symptoms including cognitive deficits, often manifested as impaired visuospatial memory. METHODS We studied cognitive performance and synaptic plasticity in a mouse model of PD, characterized by partial lesion of the dopaminergic and noradrenergic inputs to striatum and hippocampus. Sham- and 6-hydroxydopamine-lesioned mice were subjected to the novel object recognition test, and long-term potentiation was examined in the dentate gyrus and CA1 regions of the hippocampus. RESULTS Bilateral 6-hydroxydopamine lesion reduced long-term but not short-term novel object recognition and decreased long-term potentiation specifically in the dentate gyrus. These abnormalities did not depend on the loss of noradrenaline but were abolished by the antiparkinsonian drug, L-DOPA, or by SKF81297, a dopamine D1-type receptor agonist. In contrast, activation of dopamine D2-type receptors did not modify the effects produced by the lesion. Blockade of the extracellular signal-regulated kinases prevented the ability of SKF81297 to rescue novel object recognition and long-term potentiation. CONCLUSIONS These findings show that partial dopamine depletion leads to impairment of long-term recognition memory accompanied by abnormal synaptic plasticity in the dentate gyrus. They also demonstrate that activation of dopamine D1 receptors corrects these deficits, through a mechanism that requires intact extracellular signal-regulated kinases signaling.
Collapse
|
100
|
Jablonski SA, Schreiber WB, Westbrook SR, Brennan LE, Stanton ME. Determinants of novel object and location recognition during development. Behav Brain Res 2013; 256:140-50. [PMID: 23933466 DOI: 10.1016/j.bbr.2013.07.055] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/24/2013] [Accepted: 07/30/2013] [Indexed: 11/19/2022]
Abstract
In the novel object recognition (OR) paradigm, rats are placed in an arena where they encounter two sample objects during a familiarization phase. A few minutes later, they are returned to the same arena and are presented with a familiar object and a novel object. The object location recognition (OL) variant involves the same familiarization procedure but during testing one of the familiar objects is placed in a novel location. Normal adult rats are able to perform both the OR and OL tasks, as indicated by enhanced exploration of the novel vs. the familiar test item. Rats with hippocampal lesions perform the OR but not OL task indicating a role of spatial memory in OL. Recently, these tasks have been used to study the ontogeny of spatial memory but the literature has yielded conflicting results. The current experiments add to this literature by: (1) behaviorally characterizing these paradigms in postnatal day (PD) 21, 26 and 31-day-old rats; (2) examining the role of NMDA systems in OR vs. OL; and (3) investigating the effects of neonatal alcohol exposure on both tasks. Results indicate that normal-developing rats are able to perform OR and OL by PD21, with greater novelty exploration in the OR task at each age. Second, memory acquisition in the OL but not OR task requires NMDA receptor function in juvenile rats [corrected]. Lastly, neonatal alcohol exposure does not disrupt performance in either task. Implications for the ontogeny of incidental spatial learning and its disruption by developmental alcohol exposure are discussed.
Collapse
Affiliation(s)
- S A Jablonski
- Psychology Department, University of Delaware, Newark, DE 19716, USA.
| | | | | | | | | |
Collapse
|