1
|
Rossi M, Pizzorusso T. Neuroproteomics applied to the study of visual cortex plasticity Francesco. Neuroscience 2025:S0306-4522(25)00298-2. [PMID: 40258567 DOI: 10.1016/j.neuroscience.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025]
Abstract
The huge complexity of neuronal circuits arises from a temporarily overlapped influence of genetic and environmental factors (Nature and Nurture). During specific temporal windows of postnatal development, the so-called critical or sensitive periods of plasticity, the brain is particularly susceptible to the effects of experience, though this sensitivity declines with age. The most widely used experimental paradigm for studying critical periods of plasticity is the ocular dominance model in the mammalian visual cortex. Recent advancements in large-scale methodological approaches have enabled the analysis of the cellular and molecular factors regulating plasticity, highlighting the complex interaction among various metabolic and regulatory pathways. Traditionally, genomic and transcriptomic techniques have been employed to investigate the Central Nervous System in a comprehensive manner, including studies on critical period plasticity in the visual cortex. However, only have technical improvements in proteomic approaches made neuroproteomics a powerful tool for investigating both normal and pathological brain states. Despite its potential, proteomics has been underutilized in studying visual cortical plasticity. Here, we review existing studies and emphasize the importance of exploiting neuroproteomics, and of integrating with other complementary "omic" approaches, to accurately identify the true active cellular agents and ultimate mediators of brain functions.
Collapse
Affiliation(s)
- Mattia Rossi
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400 Montevideo, Uruguay.
| | - Tommaso Pizzorusso
- BIO@SNS Laboratory, Scuola Normale Superiore/Institute of Neuroscience, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
2
|
Saxena R, McNaughton BL. Bridging Neuroscience and AI: Environmental Enrichment as a model for forward knowledge transfer in continual learning. ARXIV 2025:arXiv:2405.07295v3. [PMID: 38947919 PMCID: PMC11213130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Continual learning (CL) refers to an agent's capability to learn from a continuous stream of data and transfer knowledge without forgetting old information. One crucial aspect of CL is forward transfer, i.e., improved and faster learning on a new task by leveraging information from prior knowledge. While this ability comes naturally to biological brains, it poses a significant challenge for artificial intelligence (AI). Here, we suggest that environmental enrichment (EE) can be used as a biological model for studying forward transfer, inspiring human-like AI development. EE refers to animal studies that enhance cognitive, social, motor, and sensory stimulation and is a model for what, in humans, is referred to as 'cognitive reserve'. Enriched animals show significant improvement in learning speed and performance on new tasks, typically exhibiting forward transfer. We explore anatomical, molecular, and neuronal changes post-EE and discuss how artificial neural networks (ANNs) can be used to predict neural computation changes after enriched experiences. Finally, we provide a synergistic way of combining neuroscience and AI research that paves the path toward developing AI capable of rapid and efficient new task learning.
Collapse
Affiliation(s)
- Rajat Saxena
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Bruce L McNaughton
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4 Canada
| |
Collapse
|
3
|
Zhang S, Xu W, Liu S, Xu F, Chen X, Qin H, Yao K. Anesthetic effects on electrophysiological responses across the visual pathway. Sci Rep 2024; 14:27825. [PMID: 39537872 PMCID: PMC11561267 DOI: 10.1038/s41598-024-79240-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Anesthetics are widely used in electrophysiological tests to assess retinal and visual system functions to avoid experimental errors caused by movement and stress in experimental animals. To determine the most suitable anesthetic for visual electrophysiological tests, excluding ketamine and chloral hydrate due to regulatory and side effect concerns, this study investigated the effects of ethyl carbamate (EC), avertin (AR), and pentobarbital sodium (PS) on visual signal conduction in the retina and primary visual cortex. Assessments included flash electroretinogram (FERG), pattern electroretinogram (PERG), pattern visual evoked potentials (PVEP), and flash visual evoked potentials (FVEP), FERG and FVEP were used to evaluate the responses of the retina and visual cortex to flash stimuli, respectively, while PERG and PVEP assessed responses to pattern stimuli. The research showed that AR demonstrates the least disruption to the visual signal pathway, as evidenced by consistently high characteristic peaks in the AR group across various tests. In contrast, mice given EC exhibited the lowest peak values in both FERG and FVEP, while subjects anesthetized with PS showed suppressed oscillatory potentials and PERG responses. Notably, substantial PVEP characteristic peaks were observed only in mice anesthetized with AR. Consequently, among the three anesthetics tested, AR is the most suitable for visual electrophysiological studies.
Collapse
Affiliation(s)
- Shiyao Zhang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Weihui Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Shanshan Liu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Fang Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Xiaopeng Chen
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| |
Collapse
|
4
|
Zhang N, Song B, Bai P, Du L, Chen L, Xu Y, Zeng T. Perineuronal nets' role in metabolism. Am J Physiol Endocrinol Metab 2024; 327:E411-E421. [PMID: 39140971 DOI: 10.1152/ajpendo.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Perineuronal nets (PNNs), specialized extracellular matrix (ECM) structures that envelop neurons, have recently been recognized as key players in the regulation of metabolism. This review explores the growing body of knowledge concerning PNNs and their role in metabolic control, drawing insights from recent research and relevant studies. The pivotal role of PNNs in the context of energy balance and whole body blood glucose is examined. This review also highlights novel findings, including the effects of astroglia, microglia, sex and gonadal hormones, nutritional regulation, circadian rhythms, and age on PNNs dynamics. These findings illuminate the complex and multifaceted role of PNNs in metabolic health.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Beite Song
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Peng Bai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li Du
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
5
|
Ferrer PR, Sakiyama-Elbert S. Acrylic Acid Modified Poly-ethylene Glycol Microparticles for Affinity-Based release of Insulin-Like Growth Factor-1 in Neural Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614803. [PMID: 39386667 PMCID: PMC11463357 DOI: 10.1101/2024.09.25.614803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Sustained release of bioactive molecules via affinity-based interactions presents a promising approach for controlled delivery of growth factors. Insulin-like growth factor-1 (IGF-1) has gained increased attention due to its ability to promote axonal growth in the central nervous system. In this work, we aimed to evaluate the effect of IGF-1 delivery from polyethylene-glycol diacrylate (PEG-DA) microparticles using affinity-based sustained release on neurons. We developed PEG-DA-based microparticles with varying levels of acrylic acid (AA) as a comonomer to tune their overall charge. The particles were synthesized via precipitation polymerization under UV light, yielding microparticles (MPs) with a relatively low polydispersity index. IGF-1 was incubated with the PEG-DA particles overnight, and formulations with a higher AA content resulted in higher loading efficiency and slower release rates over 4 weeks, suggesting the presence of binding interactions between the positively charged IGF-1 and negatively charged particles containing AA. The released IGF-1 was tested in dorsal root ganglion (DRG) neurite outgrowth assay and found to retain its biological activity for up to two weeks after encapsulation. Furthermore, the trophic effect of IGF-1 was tested with stem cell-derived V2a interneurons and found to have a synergistic effect when combined with neurotrophin-3 (NT3). To assess the potential of a combinatorial approach, IGF-1-releasing MPs were encapsulated within a hyaluronic acid (HA) hydrogel and showed promise as a dual delivery system. Overall, the PEG-DA MPs developed herein deliver bioactive IGF-1 for a period of weeks and hold potential to enable axonal growth of injured neurons via sustained release.
Collapse
|
6
|
Desmarchelier MR. Behavioral Development of Pediatric Exotic Pets and Practical Applications. Vet Clin North Am Exot Anim Pract 2024; 27:431-448. [PMID: 38103997 DOI: 10.1016/j.cvex.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The discovery of epigenetics and the interaction between genes and the environment have moved our understanding of how animal behavior develops from gestation to adulthood, and even throughout generations, to a new level. Studying the natural biology of exotic pets is key to providing them with a rich social and physical environment that will encourage species-specific behaviors. Combining parent-raising with appropriately timed human handling is likely to result in individuals with more resilience to stress. Using operant conditioning techniques early in life to train the animals' basic behaviors gives them control over their environment, empowering them through their social interactions.
Collapse
Affiliation(s)
- Marion R Desmarchelier
- Department of Clinical Sciences, Faculté de médecine vétérinaire, Université de Montréal, 3200 rue Sicotte, J2S 2M2 Saint-Hyacinthe, Québec, Canada.
| |
Collapse
|
7
|
Farmer AL, Lewis MH. Reduction of restricted repetitive behavior by environmental enrichment: Potential neurobiological mechanisms. Neurosci Biobehav Rev 2023; 152:105291. [PMID: 37353046 DOI: 10.1016/j.neubiorev.2023.105291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Restricted repetitive behaviors (RRB) are one of two diagnostic criteria for autism spectrum disorder and common in other neurodevelopmental and psychiatric disorders. The term restricted repetitive behavior refers to a wide variety of inflexible patterns of behavior including stereotypy, self-injury, restricted interests, insistence on sameness, and ritualistic and compulsive behavior. However, despite their prevalence in clinical populations, their underlying causes remain poorly understood hampering the development of effective treatments. Intriguingly, numerous animal studies have demonstrated that these behaviors are reduced by rearing in enriched environments (EE). Understanding the processes responsible for the attenuation of repetitive behaviors by EE should offer insights into potential therapeutic approaches, as well as shed light on the underlying neurobiology of repetitive behaviors. This review summarizes the current knowledge of the relationship between EE and RRB and discusses potential mechanisms for EE's attenuation of RRB based on the broader EE literature. Existing gaps in the literature and future directions are also discussed.
Collapse
Affiliation(s)
- Anna L Farmer
- Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Mark H Lewis
- Department of Psychology, University of Florida, Gainesville, FL, USA; Department of Psychiatry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
8
|
Bibollet-Bahena O, Tissier S, Ho-Tran S, Rojewski A, Casanova C. Enriched environment exposure during development positively impacts the structure and function of the visual cortex in mice. Sci Rep 2023; 13:7020. [PMID: 37120630 PMCID: PMC10148800 DOI: 10.1038/s41598-023-33951-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023] Open
Abstract
Optimal conditions of development have been of interest for decades, since genetics alone cannot fully explain how an individual matures. In the present study, we used optical brain imaging to investigate whether a relatively simple enrichment can positively influence the development of the visual cortex of mice. The enrichment paradigm was composed of larger cages housing multiple mice that contained several toys, hiding places, nesting material and a spinning wheel that were moved or replaced at regular intervals. We compared C57BL/6N adult mice (> P60) that had been raised either in an enriched environment (EE; n = 16) or a standard (ST; n = 12) environment from 1 week before birth to adulthood, encompassing all cortical developmental stages. Here, we report significant beneficial changes on the structure and function of the visual cortex following environmental enrichment throughout the lifespan. More specifically, retinotopic mapping through intrinsic signal optical imaging revealed that the size of the primary visual cortex was greater in mice reared in an EE compared to controls. In addition, the visual field coverage of EE mice was wider. Finally, the organization of the cortical representation of the visual field (as determined by cortical magnification) versus its eccentricity also differed between the two groups. We did not observe any significant differences between females and males within each group. Taken together, these data demonstrate specific benefits of an EE throughout development on the visual cortex, which suggests adaptation to their environmental realities.
Collapse
Affiliation(s)
- O Bibollet-Bahena
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada.
| | - S Tissier
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - S Ho-Tran
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - A Rojewski
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - C Casanova
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
9
|
Reduced expression of perineuronal nets in the normotopic somatosensory cortex of the tish rat. Brain Res 2023; 1800:148179. [PMID: 36511312 DOI: 10.1016/j.brainres.2022.148179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
The tish (telencephalic internal structural heterotopia) rat is a naturally occurring and unique model of a malformation of cortical development (MCD) arising from a sponeantous mutation in the Eml1 gene. Tish rats are characterized by a macroscopic bilateral heterotopic dysplastic cortex (HDCx) and an overlaying, intact normotopic neocortex (NNCx). These two cortices are functional and have been reported to innervate and establish connections with subcortical regions including the thalamus, resulting in a dual-cortical representation. Additionally, impaired GABAergic neurotransmission and early-onset spike wave discharge bursts have been reported in developing tish rats. Perineuronal nets (PNNs) are specialized extraceullar matrix structures that predominately surround and stabilize parvalbumin-positive (PV+) GABAergic interneurons and are essential components of the neural landscape. Here, we report a significant reduction in the average number of WFA+-PNNs in the normotopic somatosensory cortex (NSSCx) of the tish rat at two developmental time points, P16 and P35, corresponding to a decrease in the number of PV+ interneurons ensheathed by a PNN in the NSSCx. Compared with control animals, PNN expression was partially, but significantly restored following treatment with insulin-like growth factor 1 (IGF-1). These data suggest that the 'dual cortical representation' in the setting of an MCD reduces the cortical activation necessary for proper PNN expression likely contributing to the impairments in GABAergic neurotransmission and network excitability previously identified in the tish rat.
Collapse
|
10
|
An Early Enriched Experience Drives an Activated Microglial Profile at Site of Corrective Neuroplasticity in Ten-m3 Knock-Out Mice. eNeuro 2023; 10:ENEURO.0162-22.2022. [PMID: 36635245 PMCID: PMC9831145 DOI: 10.1523/eneuro.0162-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 12/15/2022] Open
Abstract
Environmental enrichment (EE) is beneficial for brain development and function, but our understanding of its capacity to drive circuit repair, the underlying mechanisms, and how this might vary with age remains limited. Ten-m3 knock-out (KO) mice exhibit a dramatic and stereotyped mistargeting of ipsilateral retinal inputs to the thalamus, resulting in visual deficits. We have recently shown a previously unexpected capacity for EE during early postnatal life (from birth for six weeks) to drive the partial elimination of miswired axonal projections, along with a recovery of visually mediated behavior, but the timeline of this repair was unclear. Here, we reveal that with just 3.5 weeks of EE from birth, Ten-m3 KOs exhibit a partial behavioral rescue, accompanied by pruning of the most profoundly miswired retinogeniculate terminals. Analysis suggests that the pruning is underway at this time point, providing an ideal opportunity to probe potential mechanisms. With the shorter EE-period, we found a localized increase in microglial density and activation profile within the identified geniculate region where corrective pruning was observed. No comparable response to EE was found in age-matched wild-type (WT) mice. These findings identify microglia as a potential mechanistic link through which EE drives the elimination of miswired neural circuits during early postnatal development. Activity driven, atypical recruitment of microglia to prune aberrant connectivity and restore function may have important therapeutic implications for neurodevelopmental disorders such as autistic spectrum disorder.
Collapse
|
11
|
Zehra Z, Khan N, Nadeem M, Siddiqui SN, von Bartheld CS, Azam M, Qamar R. Association of IGF1 polymorphisms with exotropia in a Pakistani cohort. Mol Vis 2022; 28:369-377. [PMID: 36338665 PMCID: PMC9603902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 10/04/2022] [Indexed: 06/16/2023] Open
Abstract
PURPOSE Strabismus (STBMS) is a multifactorial ocular disorder in children that leads to misalignment of the eyes. Insulin-like growth factor 1 (IGF1) has been shown to be involved in the development of extraocular muscles and myopia; however, data are limited on the genetic associations of IGF1 with STBMS in Pakistan. METHODS Two hundred seventy-four STBMS cases and 272 unaffected controls were recruited, and their DNA was extracted. Two IGF1 single nucleotide polymorphisms, rs6214 and rs5742632, were genotyped using PCR-restriction fragment length polymorphism. Univariate logistic regression analysis was performed to determine the association of these single nucleotide polymorphisms with STBMS, and the results were adjusted for age and sex. In addition, 26 extraocular muscle tissues were collected from patients with STBMS undergoing squint correction surgery, along with 3 deceased control samples. IGF1 mRNA expression was measured by quantitative PCR; the Mann-Whitney U test was applied, and fold change was calculated. Logistic regression analysis was applied to determine the association of RNA expression and fold change with genotype. RESULTS Multivariate logistic regression analysis revealed that rs5742632 (odds ratio [95% confidence interval] = 1.05[1.01-1.06], p = 0.03) is associated with STBM. Moreover, rs6214 (1.03[1.01-1.05], p = 0.03) and rs5742632 (1.09[1.04-1.11], p = 0.04) were associated with exotropia. Statistically, no significant difference in IGF1 mRNA expression in the extraocular muscles between the STBMS cases and the controls was observed. CONCLUSIONS IGF1 polymorphisms rs5742632 (A>G) and rs6214 (C>T) are plausible risk factors for the development of exotropia. However, the physiologic mechanism requires further evaluation.
Collapse
Affiliation(s)
- Zainab Zehra
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Pakistan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Netasha Khan
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Pakistan
| | - Minhal Nadeem
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Pakistan
| | | | | | - Maleeha Azam
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Pakistan
| | - Raheel Qamar
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Pakistan
- Pakistan Academy of Sciences, Islamabad, Pakistan
- Science and Technology Sector, ICESCO, Rabat, Morocco
| |
Collapse
|
12
|
Salmani N, Nozari M, Parvan M, Amini-Sardouei S, Shabani M, Khaksari M, Ezzatabadipour M. Nicotine-conditioned place preference, reversal learning, and social interaction in MK-801-induced schizophrenia model: Effects of post-weaning enriched environment. Clin Exp Pharmacol Physiol 2022; 49:871-880. [PMID: 35622536 DOI: 10.1111/1440-1681.13674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 01/22/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
Abstract
Based on the clinical observations of severe cognitive deficits in schizophrenia patients and the relationship between environmental parameters and the severity of schizophrenia symptoms, the present study investigated these parameters in an MK-801-induced schizophrenia model in rats. In addition to, it evaluated whether a post-weaning enriched environment (EE) would affect the nicotine-induced conditioned place preference (CPP) and the motor and cognitive deficits caused by MK-801 treatment. Male Wistar rat pups were injected peritoneally with MK-801 (1 mg/kg) on a daily basis between the 6th and the 10th postnatal days (P) and were exposed to either an enriched or a standard cage from P21 until the end of the experiments. The rats were evaluated in open-field and three-chamber social interaction tests. Moreover, spatial and reversal learning was assessed by the Morris water maze (MWM). Also, the animals were conditioned with 0.6 mg/kg nicotine and tested for CPP. Increased self-grooming, exploratory behavior, potentiated nicotine-CPP, and decreased social behaviors, delayed spatial learning and memory, and impaired reversal learning in the water maze were observed in the MK-801 treatment group. Housing in an EE improved cognitive and behavioral deficits associated with postnatal MK-801 treatment. The results suggested that neonatal N-methyl-D-aspartate (NMDA) receptor hypofunction may cause susceptibility to these behaviors and indicated the importance of environmental conditions in the development of schizophrenia and probably other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Neda Salmani
- Department of Psychology, Zarand Branch, Islamic Azad University, Kerman, Iran
| | - Masoumeh Nozari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Parvan
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Amini-Sardouei
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research, and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
| | - Massood Ezzatabadipour
- Anatomical Sciences Department, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
13
|
Chan J, Hao X, Liu Q, Cang J, Gu Y. Closing the Critical Period Is Required for the Maturation of Binocular Integration in Mouse Primary Visual Cortex. Front Cell Neurosci 2021; 15:749265. [PMID: 34899187 PMCID: PMC8663722 DOI: 10.3389/fncel.2021.749265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/21/2021] [Indexed: 11/25/2022] Open
Abstract
Binocular matching of orientation preference between the two eyes is a common form of binocular integration that is regarded as the basis for stereopsis. How critical period plasticity enables binocular matching under the guidance of normal visual experience has not been fully demonstrated. To investigate how critical period closure affects the binocular matching, a critical period prolonged mouse model was constructed through the administration of bumetanide, an NKCC1 transporter antagonist. Using acute in vivo extracellular recording and molecular assay, we revealed that binocular matching was transiently disrupted due to heightened plasticity after the normal critical period, together with an increase in the density of spines and synapses, and the upregulation of GluA1 expression. Diazepam (DZ)/[(R, S)-3-(2-carboxypiperazin-4-yl) propyl-1-phosphonic acid (CPP)] could reclose the extended critical period, and rescue the deficits in binocular matching. Furthermore, the extended critical period, alone, with normal visual experience is sufficient for the completion of binocular matching in amblyopic mice. Similarly, prolonging the critical period into adulthood by knocking out Nogo-66 receptor can prevent the normal maturation of binocular matching and depth perception. These results suggest that maintaining an optimal plasticity level during adolescence is most beneficial for the systemic maturation. Extending the critical period provides new clues for the maturation of binocular vision and may have critical implications for the treatment of amblyopia.
Collapse
Affiliation(s)
- Jiangping Chan
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xiangwen Hao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qiong Liu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.,School of Life Sciences, Westlake University, Hangzhou, China
| | - Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, VA, United States.,Department of Psychology, University of Virginia, Charlottesville, VA, United States
| | - Yu Gu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Gigliucci V, Teutsch J, Woodbury-Smith M, Luoni M, Busnelli M, Chini B, Banerjee A. Region-Specific KCC2 Rescue by rhIGF-1 and Oxytocin in a Mouse Model of Rett Syndrome. Cereb Cortex 2021; 32:2885-2894. [PMID: 34791112 DOI: 10.1093/cercor/bhab388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/17/2023] Open
Abstract
Rett syndrome (RTT) is characterized by dysfunction in neuronal excitation/inhibition (E/I) balance, potentially impacting seizure susceptibility via deficits in K+/Cl- cotransporter 2 (KCC2) function. Mice lacking the Methyl-CpG binding protein 2 (MeCP2) recapitulate many symptoms of RTT, and recombinant human insulin-like growth factor-1 (rhIGF-1) restores KCC2 expression and E/I balance in MeCP2 KO mice. However, clinical trial outcomes of rhIGF-1 in RTT have been variable, and increasing its therapeutic efficacy is highly desirable. To this end, the neuropeptide oxytocin (OXT) is promising, as it also critically modulates KCC2 function during early postnatal development. We measured basal KCC2 expression levels in MeCP2 KO mice and identified 3 key frontal brain regions showing KCC2 alterations in young adult mice, but not in postnatal P10 animals. We hypothesized that deficits in an IGF-1/OXT signaling crosstalk modulating KCC2 may occur in RTT during postnatal development. Consistently, we detected alterations of IGF-1 receptor and OXT receptor levels in those brain areas. rhIGF-1 and OXT treatments in KO mice rescued KCC2 expression in a region-specific and complementary manner. These results suggest that region-selective combinatorial pharmacotherapeutic strategies could be most effective at normalizing E/I balance in key brain regions subtending the RTT pathophysiology.
Collapse
Affiliation(s)
| | - Jasper Teutsch
- Neuroscience Theme, Biosciences Institute, Newcastle University, United Kingdom.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Marc Woodbury-Smith
- Neuroscience Theme, Biosciences Institute, Newcastle University, United Kingdom
| | - Mirko Luoni
- Stem Cells and Neurogenesis Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Marta Busnelli
- Institute of Neuroscience, CNR, Milan, Italy.,NeuroMi Milan Center for Neuroscience, Milan, Italy
| | - Bice Chini
- Institute of Neuroscience, CNR, Milan, Italy.,NeuroMi Milan Center for Neuroscience, Milan, Italy
| | - Abhishek Banerjee
- Neuroscience Theme, Biosciences Institute, Newcastle University, United Kingdom.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Consorti A, Di Marco I, Sansevero G. Physical Exercise Modulates Brain Physiology Through a Network of Long- and Short-Range Cellular Interactions. Front Mol Neurosci 2021; 14:710303. [PMID: 34489641 PMCID: PMC8417110 DOI: 10.3389/fnmol.2021.710303] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
In the last decades, the effects of sedentary lifestyles have emerged as a critical aspect of modern society. Interestingly, recent evidence demonstrated that physical exercise plays an important role not only in maintaining peripheral health but also in the regulation of central nervous system function. Many studies have shown that physical exercise promotes the release of molecules, involved in neuronal survival, differentiation, plasticity and neurogenesis, from several peripheral organs. Thus, aerobic exercise has emerged as an intriguing tool that, on one hand, could serve as a therapeutic protocol for diseases of the nervous system, and on the other hand, could help to unravel potential molecular targets for pharmacological approaches. In the present review, we will summarize the cellular interactions that mediate the effects of physical exercise on brain health, starting from the factors released in myocytes during muscle contraction to the cellular pathways that regulate higher cognitive functions, in both health and disease.
Collapse
Affiliation(s)
- Alan Consorti
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- NEUROFARBA, University of Florence, Florence, Italy
| | | | | |
Collapse
|
16
|
Carbone BE, Abouleish M, Watters KE, Vogel S, Ribic A, Schroeder OHU, Bader BM, Biederer T. Synaptic Connectivity and Cortical Maturation Are Promoted by the ω-3 Fatty Acid Docosahexaenoic Acid. Cereb Cortex 2021; 30:226-240. [PMID: 31034037 DOI: 10.1093/cercor/bhz083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/20/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022] Open
Abstract
Brain development is likely impacted by micronutrients. This is supported by the effects of the ω-3 fatty acid docosahexaenoic acid (DHA) during early neuronal differentiation, when it increases neurite growth. Aiming to delineate DHA roles in postnatal stages, we selected the visual cortex due to its stereotypic maturation. Immunohistochemistry showed that young mice that received dietary DHA from birth exhibited more abundant presynaptic and postsynaptic specializations. DHA also increased density and size of synapses in a dose-dependent manner in cultured neurons. In addition, dendritic arbors of neurons treated with DHA were more complex. In agreement with improved connectivity, DHA enhanced physiological parameters of network maturation in vitro, including bursting strength and oscillatory behavior. Aiming to analyze functional maturation of the cortex, we performed in vivo electrophysiological recordings from awake mice to measure responses to patterned visual inputs. Dietary DHA robustly promoted the developmental increase in visual acuity, without altering light sensitivity. The visual acuity of DHA-supplemented animals continued to improve even after their cortex had matured and DHA abolished the acuity plateau. Our findings show that the ω-3 fatty acid DHA promotes synaptic connectivity and cortical processing. These results provide evidence that micronutrients can support the maturation of neuronal networks.
Collapse
Affiliation(s)
- Beatrice E Carbone
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Malik Abouleish
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Katherine E Watters
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Seth Vogel
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Adema Ribic
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | | | | | - Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
17
|
Transplantation of Neural Precursors Derived from Induced Pluripotent Cells Preserve Perineuronal Nets and Stimulate Neural Plasticity in ALS Rats. Int J Mol Sci 2020; 21:ijms21249593. [PMID: 33339362 PMCID: PMC7766921 DOI: 10.3390/ijms21249593] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/05/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022] Open
Abstract
A promising therapeutic strategy for amyotrophic lateral sclerosis (ALS) treatment is stem cell therapy. Neural progenitors derived from induced pluripotent cells (NP-iPS) might rescue or replace dying motoneurons (MNs). However, the mechanisms responsible for the beneficial effect are not fully understood. The aim here was to investigate the mechanism by studying the effect of intraspinally injected NP-iPS into asymptomatic and early symptomatic superoxide dismutase (SOD)1G93A transgenic rats. Prior to transplantation, NP-iPS were characterized in vitro for their ability to differentiate into a neuronal phenotype. Motor functions were tested in all animals, and the tissue was analyzed by immunohistochemistry, qPCR, and Western blot. NP-iPS transplantation significantly preserved MNs, slowed disease progression, and extended the survival of all treated animals. The dysregulation of spinal chondroitin sulfate proteoglycans was observed in SOD1G93A rats at the terminal stage. NP-iPS application led to normalized host genes expression (versican, has-1, tenascin-R, ngf, igf-1, bdnf, bax, bcl-2, and casp-3) and the protection of perineuronal nets around the preserved MNs. In the host spinal cord, transplanted cells remained as progenitors, many in contact with MNs, but they did not differentiate. The findings suggest that NP-iPS demonstrate neuroprotective properties by regulating local gene expression and regulate plasticity by modulating the central nervous system (CNS) extracellular matrix such as perineuronal nets (PNNs).
Collapse
|
18
|
Reh R, Williams LJ, Todd RM, Ward LM. Warped rhythms: Epileptic activity during critical periods disrupts the development of neural networks for human communication. Behav Brain Res 2020; 399:113016. [PMID: 33212087 DOI: 10.1016/j.bbr.2020.113016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/27/2022]
Abstract
It is well established that temporal lobe epilepsy-the most common and well-studied form of epilepsy-can impair communication by disrupting social-emotional and language functions. In pediatric epilepsy, where seizures co-occur with the development of critical brain networks, age of onset matters: The earlier in life seizures begin, the worse the disruption in network establishment, resulting in academic hardship and social isolation. Yet, little is known about the processes by which epileptic activity disrupts developing human brain networks. Here we take a synthetic perspective-reviewing a range of research spanning studies on molecular and oscillatory processes to those on the development of large-scale functional networks-in support of a novel model of how such networks can be disrupted by epilepsy. We seek to bridge the gap between research on molecular processes, on the development of human brain circuitry, and on clinical outcomes to propose a model of how epileptic activity disrupts brain development.
Collapse
Affiliation(s)
- Rebecca Reh
- University of British Columbia, Department of Psychology, 2136 West Mall, Vancouver BC V6T 1Z4, Canada
| | - Lynne J Williams
- BC Children's Hospital MRI Research Facility, 4480 Oak Street, Vancouver, BC V6H 0B3, Canada
| | - Rebecca M Todd
- University of British Columbia, Department of Psychology, 2136 West Mall, Vancouver BC V6T 1Z4, Canada; University of British Columbia, Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Lawrence M Ward
- University of British Columbia, Department of Psychology, 2136 West Mall, Vancouver BC V6T 1Z4, Canada; University of British Columbia, Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
19
|
Snell-Rood E, Snell-Rood C. The developmental support hypothesis: adaptive plasticity in neural development in response to cues of social support. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190491. [PMID: 32475336 PMCID: PMC7293157 DOI: 10.1098/rstb.2019.0491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Across mammals, cues of developmental support, such as touching, licking or attentiveness, stimulate neural development, behavioural exploration and even overall body growth. Why should such fitness-related traits be so sensitive to developmental conditions? Here, we review what we term the 'developmental support hypothesis', a potential adaptive explanation of this plasticity. Neural development can be a costly process, in terms of time, energy and exposure. However, environmental variability may sometimes compromise parental care during this costly developmental period. We propose this environmental variation has led to the evolution of adaptive plasticity of neural and behavioural development in response to cues of developmental support, where neural development is stimulated in conditions that support associated costs. When parental care is compromised, offspring grow less and adopt a more resilient and stress-responsive strategy, improving their chances of survival in difficult conditions, similar to existing ideas on the adaptive value of early-life programming of stress. The developmental support hypothesis suggests new research directions, such as testing the adaptive value of reduced neural growth and metabolism in stressful conditions, and expanding the range of potential cues animals may attend to as indicators of developmental support. Considering evolutionary and ecologically appropriate cues of social support also has implications for promoting healthy neural development in humans. This article is part of the theme issue 'Life history and learning: how childhood, caregiving and old age shape cognition and culture in humans and other animals'.
Collapse
Affiliation(s)
- Emilie Snell-Rood
- Department of Ecology, Evolution and Behavior, University of Minnesota, 1479 Gortner Avenue, Gortner 140, St Paul, MN 55108, USA
| | - Claire Snell-Rood
- School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
20
|
Zheng JJ, Zou R, Huang S, Song TJ, Yu X. Enriched Environment Rearing from Birth Reduced Anxiety, Improved Learning and Memory, and Promoted Social Interactions in Adult Male Mice. Neuroscience 2020; 442:138-150. [PMID: 32652178 DOI: 10.1016/j.neuroscience.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/11/2020] [Accepted: 07/02/2020] [Indexed: 12/21/2022]
Abstract
Rearing rodents in an enriched environment (EE), with increased sensory stimulations and social interactions, is a well-established model for naturally increasing neural activity. It is well-known that EE-rearing of rodents from adolescence or during adulthood leads to extensive biochemical, morphological, electrophysiological and behavioral changes. Here, we examine the effects of EE-rearing from birth on adult behavior. Through a battery of assays, we found that mice EE-reared from birth had better acquisition and consolidation of memory, in both aversive-based fear conditioning and reward-based contextual association tasks. Moreover, EE-reared mice showed reduced anxiety in novel environments and enhanced social interactions. Together, these results demonstrated that EE-rearing from birth significantly improved motor ability, learning and memory and sociability, while reducing anxiety. A better understanding of how early environmental influences affect behavior is not only important for understanding neural circuit wiring, but also provides insight into developing more effective intervention programs for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jing-Jing Zheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rong Zou
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shajin Huang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tian-Jia Song
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China.
| | - Xiang Yu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China.
| |
Collapse
|
21
|
Luke MPS, Brown RE, Clarke DB. Polysialylated - neural cell adhesion molecule (PSA-NCAM) promotes recovery of vision after the critical period. Mol Cell Neurosci 2020; 107:103527. [PMID: 32634575 DOI: 10.1016/j.mcn.2020.103527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 06/05/2020] [Accepted: 06/29/2020] [Indexed: 01/19/2023] Open
Abstract
Vision loss has long since been considered irreversible after a critical period; however, there is potential to restore limited vision, even in adulthood. This phenomenon is particularly pronounced following complete loss of vision in the dominant eye. Adult neural cell adhesion molecule (NCAM) knockout mice have an age-related impairment of visual acuity. The underlying cause of early deterioration in visual function remains unknown. Polysialylated (PSA) NCAM is involved in different forms of neural plasticity in the adult brain, raising the possibility that NCAM plays a role in the plasticity of the visual cortex, and therefore, in visual ability. Here, we examined whether PSA-NCAM is required for visual cortical plasticity in adult C57Bl/6J mice following deafferentation and long-term monocular deprivation. Our results show that elevated PSA in the contralateral visual cortex of the reopened eye is accompanied by changes in other markers of neural plasticity: increased brain-derived neurotrophic factor (BDNF) levels and degradation of perineuronal nets (PNNs). The removal of PSA-NCAM in the visual cortex of these mice reduced BDNF expression, decreased PNN degradation, and resulted in impaired recovery of visual acuity after optic nerve transection and chronic monocular deprivation. Collectively, our results demonstrate that PSA-NCAM is necessary for the reactivation of visual cortical plasticity and recovery of visual function in adult mice. It also offers a potential molecular target for the therapeutic treatment of cortically based visual impairments.
Collapse
Affiliation(s)
- Margaret Po-Shan Luke
- Department of Medical Neuroscience, Dalhousie University, Life Science Research Institute, 1348 Summer Street, Halifax B3H 4R2, NS, Canada.
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Life Science Centre, 1355 Oxford Street, PO Box 15000, Halifax B3H 4R2, NS, Canada.
| | - David B Clarke
- Departments of Surgery (Neurosurgery), Medical Neuroscience, and Ophthalmology & Visual Sciences, Dalhousie University, Life Science Research Institute, 1348 Summer Street, Halifax B3H 4R2, NS, Canada.
| |
Collapse
|
22
|
Yang L, Yang Y, Yuan J, Sun Y, Dai J, Su B. Transcriptomic Landscape of von Economo Neurons in Human Anterior Cingulate Cortex Revealed by Microdissected-Cell RNA Sequencing. Cereb Cortex 2020; 29:838-851. [PMID: 30535007 PMCID: PMC6319179 DOI: 10.1093/cercor/bhy286] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Indexed: 01/19/2023] Open
Abstract
The von Economo neurons (VENs) are specialized large bipolar projection neurons with restricted distribution in the human brain, and they are far more abundant in humans than in non-human primates. However, VEN functions remain elusive due to the difficulty of isolating VENs and dissecting their connections in the brain. Here, we combined laser-capture-microdissection with RNA sequencing to describe the transcriptomic profile of VENs from human anterior cingulate cortex (ACC). Using pyramidal neurons as reference cells, we identified 344 genes with VEN-associated expression differences, including 215 higher and 129 lower expression genes. Functional enrichment and protein–protein interaction network analyses showed that these genes with VEN-associated expression differences are involved in VEN morphogenesis and functions, such as dendrite branching and axon myelination, and many of them are associated with human social-emotional disorders. With the use of in situ hybridization and immunohistochemistry assays, we validated four novel VEN markers (VAT1L, CHST8, LYPD1, and SULF2). Collectively, we generated a full-spectrum expression profile of VENs from human ACC, greatly enlarging the pool of genes with VEN-associated expression differences that can help researchers to understand the role of VENs in normal and disordered human brains.
Collapse
Affiliation(s)
- Lixin Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Yandong Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jiamiao Yuan
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yan Sun
- Chinese Brain Bank Center, South-Central University for Nationalities, Wuhan, China
| | - Jiapei Dai
- Chinese Brain Bank Center, South-Central University for Nationalities, Wuhan, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
23
|
Luo Y, Liu Z, Luo S, Wang X, Tao L. The developmental and experience-dependent expression of IGF-2 in mice visual cortex. Neurosci Lett 2020; 721:134828. [PMID: 32044392 DOI: 10.1016/j.neulet.2020.134828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 12/21/2019] [Accepted: 02/06/2020] [Indexed: 10/25/2022]
Abstract
The circuitry associated with the visual cortex is particularly sensitive to experiences during the early stages of life, which are collectively known as critical periods. Critical period of ocular dominance plasticity is regulated by both environmental and genetic factors. Previous studies demonstrated that IGF-1 significantly influenced the regulation of visual cortex synaptic plasticity. IGF-2 can reportedly regulate synapse formation, dendritic spine maturation, and memory consolidation in rodents. Association between IGF-2 and the regulation of visual cortex synaptic plasticity remains unclear. Here, we first aimed to elucidate the normal expression patterns of IGF-2 and its laminar expression pattern during the process of visual cortex development in mice. This confirmed that IGF-2 may influence the regulation of ocular dominance plasticity in mice. We further elucidated the role of IGF-2 in the regulation of visual cortex synaptic plasticity by examining the effect of monocular deprivation (MD) on IGF-2 expression in the visual cortex. Interestingly, we observed that MD remarkably reduced IGF-2 expression in the visual cortex. Rodents reared in an enriched environment, with enhanced sensory, motor, and social experiences, were capable of effectively accelerating the development of the visual system and could restore normal visual acuity. Although the enriched environment facilitated the restoration of normal visual acuity in the MD mice, IGF-2 expression levels in the visual cortex remained unchanged. Therefore, we considered the possibility that IGF-2 may have a different role with regard to the modulation of plasticity in the visual cortex of the mice, which we aim to study in the future.
Collapse
Affiliation(s)
- Yulin Luo
- Department of Ophthalmology, Hunan Children's Hospital, Changsha 410007, China.
| | - Zhenghai Liu
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang 421001, China
| | - Shishi Luo
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang 421001, China
| | - Xilang Wang
- Department of Ophthalmology, Hunan Children's Hospital, Changsha 410007, China
| | - Lijuan Tao
- Department of Ophthalmology, Hunan Children's Hospital, Changsha 410007, China
| |
Collapse
|
24
|
Blok J, Black DA, Petersen J, Sawatari A, Leamey CA. Environmental Enrichment Rescues Visually-Mediated Behavior in Ten-m3 Knockout Mice During an Early Critical Period. Front Behav Neurosci 2020; 14:22. [PMID: 32158383 PMCID: PMC7052109 DOI: 10.3389/fnbeh.2020.00022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 01/31/2020] [Indexed: 11/13/2022] Open
Abstract
Environmental enrichment (EE) has been shown to promote neural plasticity. Its capacity to induce functional repair in models which exhibit profound sensory deficits due to aberrant axonal guidance has not been well-characterized. Ten-m3 knockout (KO) mice exhibit a highly-stereotyped miswiring of ipsilateral retinogeniculate axons and associated profound deficits in binocularly-mediated visual behavior. We determined whether, and when, EE can drive functional recovery by analyzing Ten-m3 KO and wildtype (WT) mice that were enriched for 6 weeks from adulthood, weaning or birth in comparison to standard-housed controls. EE initiated from birth, but not later, rescued the response of Ten-m3 KOs to the "looming" stimulus (expanding disc in dorsal visual field), suggesting improved visual function. EE can thus induce recovery of visual behavior, but only during an early developmentally-restricted time-window.
Collapse
Affiliation(s)
- James Blok
- Department of Physiology, School of Medical Sciences and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Dylan A Black
- Department of Physiology, School of Medical Sciences and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Justin Petersen
- Department of Physiology, School of Medical Sciences and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Atomu Sawatari
- Department of Physiology, School of Medical Sciences and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Catherine A Leamey
- Department of Physiology, School of Medical Sciences and Bosch Institute, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
25
|
Insulin-like growth factor 1 is related to the expression of plumage traits in a passerine species. Behav Ecol Sociobiol 2020. [DOI: 10.1007/s00265-020-2821-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Abstract
Avian plumage colors and ornaments are excellent models to study the endocrine mechanisms linking sexually selected traits and individual parameters of quality and condition. Insulin-like growth factor 1 (IGF-1) is an evolutionarily highly conserved peptide hormone. Its regulatory role in cell proliferation and differentiation and its high sensitivity to the nutritional state of individuals suggest it as an interesting candidate, possibly providing a link between body condition and individual capacity to grow elaborated ornamental features. We investigated whether IGF-1 levels during molting correlate with the expression of multiple ornaments in a sexually dichromatic passerine species, the bearded reedling (Panurus biarmicus). We collected blood samples of males and females shortly before the molting completed and measured the size and colors of ornamental traits. Our results indicate that in males, structural plumage colors, the size of the melanin-based ornament (beard), and tail length are independent traits. IGF-1 levels are associated with the length of the tail and the expression of male structural plumage components (UV coloration), but not the melanin-based ornament. In females, plumage color and tail length were independent traits, which were not related to IGF-1 levels. To the best of our knowledge, this study provides the first evidence that IGF-1 could play a role in the development of secondary sexual characters in a bird species.
Significance statement
IGF-1 is an evolutionarily highly conserved peptide hormone, which recently entered the center stage of research enquiry in evolutionary biology. It is considered as one of the key factors shaping individual life histories, but little is known about its effects on sexually selected traits. We investigated whether IGF-1 levels during molting predict the elaboration of multiple ornamental plumage traits in male and female bearded reedlings (Panurus biarmicus). Our results indicate that higher IGF-1 levels had positive effects on male structural plumage colors and tail feather length. This is the first study, bringing indication for a potential role of IGF-1 in the expression of plumage ornaments in a bird species. Our findings suggest that IGF-1 might serve as an ideal candidate to study the mechanisms linking condition and the capacity to develop sexually selected ornaments.
Collapse
|
26
|
Insulin-like growth factor-1 inhibits spreading depression-induced trigeminal calcitonin gene related peptide, oxidative stress & neuronal activation in rat. Brain Res 2020; 1732:146673. [PMID: 31978377 DOI: 10.1016/j.brainres.2020.146673] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 11/21/2022]
Abstract
Migraineurs can show brain hyperexcitability and oxidative stress that may promote headache. Since hyperexcitability can enhance oxidative stress which promotes hyperexcitability, ending this feed-back loop may reduce migraine. Neocortical spreading depression, an animal model of migraine begins with hyperexcitability and triggers oxidative stress in the neocortical area involved and in the trigeminal system, which is important to pain pathway nociceptive activation in migraine. Additionally, oxidative stress causes increased trigeminal ganglion calcitonin gene-related peptide release and oxidative stress can reduce spreading depression threshold. Insulin-like growth factor-1 significantly protects against spreading depression in vitro by reducing oxidative stress and it is effective against spreading depression after intranasal delivery to animals. Here, we used adult male rats and extend this work to study the trigeminal system where insulin-like growth factor-1 receptors are highly expressed. Recurrent neocortical spreading depression significantly increased surrogate markers of trigeminal activation - immunostaining for trigeminal ganglion oxidative stress, calcitonin gene related peptide levels and c-fos in the trigeminocervical complex versus sham. These effects were significantly reduced by intranasal delivery of insulin-like growth factor-1 a day before recurrent neocortical spreading depression. Furthermore, intranasal treatment with insulin-like growth factor-1 significantly reduced naïve levels of trigeminal ganglion calcitonin gene related peptide versus sham with no impact on blood glucose levels. Intranasal delivery of insulin-like growth factor-1 not only mitigates neocortical spreading depression, a cause of migraine hyperexcitability modeled in animals, but also when neocortical spreading depression is triggered by supra-threshold stimuli, insulin-like growth factor-1 effectively reduces nociceptive activation in the trigeminal system.
Collapse
|
27
|
Environmental Enrichment Partially Repairs Subcortical Mapping Errors in Ten-m3 Knock-Out Mice during an Early Critical Period. eNeuro 2019; 6:ENEURO.0478-18.2019. [PMID: 31767573 PMCID: PMC6901682 DOI: 10.1523/eneuro.0478-18.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 09/23/2019] [Accepted: 10/19/2019] [Indexed: 11/21/2022] Open
Abstract
Environmental enrichment (EE) has been shown to improve neural function via the regulation of cortical plasticity. Its capacity to induce functional and/or anatomical repair of miswired circuits is unknown. Ten-m3 knock-out (KO) mice exhibit a highly stereotyped and profound miswiring of ipsilateral retinogeniculate axons and associated deficits in binocularly-mediated visual behavior. We determined whether, and when, EE can drive the repair of subcortical wiring deficits by analyzing Ten-m3 KO and wild-type (WT) mice that were enriched for six weeks from adulthood, weaning or birth in comparison to standard-housed (SE) controls. Six weeks of EE initiated from birth, but not later, induced a significant reduction in the area occupied by ipsilateral retinogeniculate terminals in KOs. No EE-induced correction of mistargeted axons was observed at postnatal day (P)7, indicating that this intervention impacts pruning rather than initial targeting of axons. This reduction was most prominent in the ventrolateral region of the dorsal lateral geniculate nucleus (dLGN), suggesting a preferential pruning of the most profoundly mistargeted axons. EE can thus partially repair a specific, subcortical axonal wiring deficit, but only during an early, developmentally-restricted time window.
Collapse
|
28
|
O'Connor AM, Burton TJ, Mansuri H, Hand GR, Leamey CA, Sawatari A. Environmental Enrichment From Birth Impacts Parvalbumin Expressing Cells and Wisteria Floribunda Agglutinin Labelled Peri-Neuronal Nets Within the Developing Murine Striatum. Front Neuroanat 2019; 13:90. [PMID: 31708753 PMCID: PMC6821641 DOI: 10.3389/fnana.2019.00090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 10/01/2019] [Indexed: 11/13/2022] Open
Abstract
Environmental enrichment can dramatically affect both the development and function of neural circuits. This is accomplished, at least in part, by the regulation of inhibitory cellular networks and related extracellular matrix glycoprotein structures known as perineuronal nets. The degree to which enhanced housing can influence brain areas involved in the planning and execution of actions is not well known. We examined the effect of enriching mice from birth on parvalbumin expression and perineuronal net formation in developing and adult striatum. This input nucleus of the basal ganglia consists of topographically discernible regions that serve different functions, providing a means of simultaneously examining the influence of environmental factors on discrete, but related networks. Greater densities of striatal parvalbumin positive cells and wisteria floribunda agglutinin labelled perineuronal nets were present in enriched pups during the second postnatal week, primarily within the lateral portion of the nucleus. Housing conditions continued to have an impact into adulthood, with enriched mice exhibiting higher parvalbumin positive cell densities in both medial and lateral striatum. Curiously, no differences due to housing conditions were detected in striatal perineuronal net densities of mature animals. The degree of overlap between striatal parvalbumin expression and perineuronal net formation was also increased, suggesting that heightened neural activity associated with enrichment may have contributed to greater engagement of networks affiliated with cells that express the calcium binding protein. Brain derived neurotrophic factor, an important regulator of inhibitory network maturation, is also subtly, but significantly affected within the striatum of enriched cohorts. Together, these findings suggest that environmental enrichment can exert cell specific effects within different divisions of an area vital for the regulation of action.
Collapse
Affiliation(s)
- Angela May O'Connor
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Thomas Joseph Burton
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Hannan Mansuri
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gabriel Rhys Hand
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Catherine Anne Leamey
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Atomu Sawatari
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
29
|
Santi A, Genis L, Torres Aleman I. A Coordinated Action of Blood-Borne and Brain Insulin-Like Growth Factor I in the Response to Traumatic Brain Injury. Cereb Cortex 2019; 28:2007-2014. [PMID: 28449086 DOI: 10.1093/cercor/bhx106] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/11/2017] [Indexed: 01/20/2023] Open
Abstract
In response to injury, the brain produces different neuroprotective molecules, such as insulin-like growth factor I (IGF-I). However, IGF-I is also taken up by the brain from the circulation in response to physiological stimuli. Herein, we analyzed in mice the relative contribution of circulating and locally produced IGF-I to increased brain IGF-I levels after insult. Traumatic brain injury (TBI) induced by a controlled impact resulted in increased IGF-I levels in the vicinity of the lesion, but mice with low serum IGF-I showed significantly lower increases. Indeed, in normal mice, peripheral IGF-I accumulated at the lesion site after injury, and at the same time serum IGF-I levels decreased. Collectively, these data suggest that serum IGF-I enter into the brain after TBI and contributes to increased brain IGF-I levels at the injury site. This connection between central and circulating IGF-I provides an amenable route for treatment, as subcutaneous administration of IGF-I to TBI mice led to functional recovery. These latter results add further support to the use of systemic IGF-I or its mimetics for treatment of brain injuries.
Collapse
Affiliation(s)
- A Santi
- Cajal Institute, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain.,Ciberned, C/ Valderrebollo 5, 28031 Madrid, Spain
| | - L Genis
- Cajal Institute, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain.,Ciberned, C/ Valderrebollo 5, 28031 Madrid, Spain
| | - I Torres Aleman
- Cajal Institute, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain.,Ciberned, C/ Valderrebollo 5, 28031 Madrid, Spain
| |
Collapse
|
30
|
Liu E, Zhou Q, Xie AJ, Li M, Zhang S, Huang H, Liuyang Z, Wang Y, Liu B, Li X, Sun D, Wei Y, Wang X, Wang Q, Ke D, Yang X, Yang Y, Wang JZ. Enriched gestation activates the IGF pathway to evoke embryo-adult benefits to prevent Alzheimer's disease. Transl Neurodegener 2019; 8:8. [PMID: 30867903 PMCID: PMC6399936 DOI: 10.1186/s40035-019-0149-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/15/2019] [Indexed: 12/13/2022] Open
Abstract
Background Building brain reserves before dementia onset could represent a promising strategy to prevent Alzheimer's disease (AD), while how to initiate early cognitive stimulation is unclear. Given that the immature brain is more sensitive to environmental stimuli and that brain dynamics decrease with ageing, we reasoned that it would be effective to initiate cognitive stimulation against AD as early as the fetal period. Methods After conception, maternal AD transgenic mice (3 × Tg AD) were exposed to gestational environment enrichment (GEE) until the day of delivery. The cognitive capacity of the offspring was assessed by the Morris water maze and contextual fear-conditioning tests when the offspring were raised in a standard environment to 7 months of age. Western blotting, immunohistochemistry, real-time PCR, immunoprecipitation, chromatin immunoprecipitation (ChIP) assay, electrophysiology, Golgi staining, activity assays and sandwich ELISA were employed to gain insight into the mechanisms underlying the beneficial effects of GEE on embryos and 7-10-month-old adult offspring. Results We found that GEE markedly preserved synaptic plasticity and memory capacity with amelioration of hallmark pathologies in 7-10-m-old AD offspring. The beneficial effects of GEE were accompanied by global histone hyperacetylation, including those at bdnf promoter-binding regions, with robust BDNF mRNA and protein expression in both embryo and progeny hippocampus. GEE increased insulin-like growth factor 1 (IGF1) and activated its receptor (IGF1R), which phosphorylates Ca2+/calmodulin-dependent kinase IV (CaMKIV) at tyrosine sites and triggers its nuclear translocation, subsequently upregulating histone acetyltransferase (HAT) and BDNF transcription. The upregulation of IGF1 mimicked the effects of GEE, while IGF1R or HAT inhibition during pregnancy abolished the GEE-induced CaMKIV-dependent histone hyperacetylation and BDNF upregulation. Conclusions These findings suggest that activation of IGF1R/CaMKIV/HAT/BDNF signaling by gestational environment enrichment may serve as a promising strategy to delay AD progression.
Collapse
Affiliation(s)
- Enjie Liu
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China.,4Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Qiuzhi Zhou
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Ao-Ji Xie
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Mengzhu Li
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Shujuan Zhang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Hezhou Huang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Zhenyu Liuyang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yali Wang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Bingjin Liu
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xiaoguang Li
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Dongsheng Sun
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yuping Wei
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xiaochuan Wang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Qun Wang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Dan Ke
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Centre for Disease Control and Prevention, 8 Longyuan Road, Shenzhen, 518055 China
| | - Ying Yang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Jian-Zhi Wang
- 1Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China.,2Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000 China
| |
Collapse
|
31
|
Durán-Carabali L, Arcego D, Sanches E, Odorcyk F, Marques M, Tosta A, Reichert L, Carvalho A, Dalmaz C, Netto C. Preventive and therapeutic effects of environmental enrichment in Wistar rats submitted to neonatal hypoxia-ischemia. Behav Brain Res 2019; 359:485-497. [DOI: 10.1016/j.bbr.2018.11.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/14/2018] [Accepted: 11/24/2018] [Indexed: 12/27/2022]
|
32
|
Dobolyi A, Lékó AH. The insulin-like growth factor-1 system in the adult mammalian brain and its implications in central maternal adaptation. Front Neuroendocrinol 2019; 52:181-194. [PMID: 30552909 DOI: 10.1016/j.yfrne.2018.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/04/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Our knowledge on the bioavailability and actions of insulin-like growth factor-1 (IGF-1) has markedly expanded in recent years as novel mechanisms were discovered on IGF binding proteins (IGFBPs) and their ability to release IGF-1. The new discoveries allowed a better understanding of the endogenous physiological actions of IGF-1 and also its applicability in therapeutics. The focus of the present review is to summarize novel findings on the neuronal, neuroendocrine and neuroplastic actions of IGF-1 in the adult brain. As most of the new regulatory mechanisms were described in the periphery, their implications on brain IGF system will also be covered. In addition, novel findings on the effects of IGF-1 on lactation and maternal behavior are described. Based on the enormous neuroplastic changes related to the peripartum period, IGF-1 has great but largely unexplored potential in maternal adaptation of the brain, which is highlighted in the present review.
Collapse
Affiliation(s)
- Arpád Dobolyi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary.
| | - András H Lékó
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary; Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary; Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
33
|
Synaptic and circuit development of the primary sensory cortex. Exp Mol Med 2018; 50:1-9. [PMID: 29628505 PMCID: PMC5938038 DOI: 10.1038/s12276-018-0029-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 01/06/2023] Open
Abstract
Animals, including humans, optimize their primary sensory cortex through the use of input signals, which allow them to adapt to the external environment and survive. The time window at the beginning of life in which external input signals are connected sensitively and strongly to neural circuit optimization is called the critical period. The critical period has attracted the attention of many neuroscientists due to the rapid activity-/experience-dependent circuit development that occurs, which is clearly differentiated from other developmental time periods and brain areas. This process involves various types of GABAergic inhibitory neurons, the extracellular matrix, neuromodulators, transcription factors, and neurodevelopmental factors. In this review, I discuss recent progress regarding the biological nature of the critical period that contribute to a better understanding of brain development.
Collapse
|
34
|
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
|
35
|
A Systematic Look at Environmental Modulation and Its Impact in Brain Development. Trends Neurosci 2018; 41:4-17. [DOI: 10.1016/j.tins.2017.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/11/2017] [Accepted: 10/18/2017] [Indexed: 11/19/2022]
|
36
|
Grinberg YY, Zitzow LA, Kraig RP. Intranasally administered IGF-1 inhibits spreading depression in vivo. Brain Res 2017; 1677:47-57. [PMID: 28951235 DOI: 10.1016/j.brainres.2017.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022]
Abstract
Spreading depression (SD) is a wave of cellular depolarization that travels slowly through susceptible gray matter brain areas. SD is the most likely cause of migraine aura and perhaps migraine pain, and is a well-accepted animal model of migraine. Identification of therapeutics that can prevent SD may have clinical relevance toward migraine treatment. Here we show that insulin-like growth factor-1 (IGF-1) significantly inhibited neocortical SD in vivo after intranasal delivery to rats. A single dose of IGF-1 inhibited SD within an hour, and continued to protect for at least seven days thereafter. A two-week course of IGF-1, administered every third day, further decreased SD susceptibility and showed no aberrant effects on glial activation, nasal mucosa, or serum markers of toxicity. SD begets SD in vitro by mechanisms that involve microglial activation. We add to this relationship by showing that recurrent SD in vivo increased susceptibility to subsequent SD, and that intervention with IGF-1 significantly interrupted this pathology. These findings support nasal administration of IGF-1 as a novel intervention capable of mitigating SD susceptibility, and as a result, potentially migraine.
Collapse
Affiliation(s)
- Yelena Y Grinberg
- Department of Neurology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, United States
| | - Lois A Zitzow
- Animal Resources Center, Department of Surgery, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, United States
| | - Richard P Kraig
- Department of Neurology, The University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637-1470, United States.
| |
Collapse
|
37
|
Marchetto MC, Belinson H, Tian Y, Freitas BC, Fu C, Vadodaria K, Beltrao-Braga P, Trujillo CA, Mendes AP, Padmanabhan K, Nunez Y, Ou J, Ghosh H, Wright R, Brennand K, Pierce K, Eichenfield L, Pramparo T, Eyler L, Barnes CC, Courchesne E, Geschwind DH, Gage FH, Wynshaw-Boris A, Muotri AR. Altered proliferation and networks in neural cells derived from idiopathic autistic individuals. Mol Psychiatry 2017; 22:820-835. [PMID: 27378147 PMCID: PMC5215991 DOI: 10.1038/mp.2016.95] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/29/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorders (ASD) are common, complex and heterogeneous neurodevelopmental disorders. Cellular and molecular mechanisms responsible for ASD pathogenesis have been proposed based on genetic studies, brain pathology and imaging, but a major impediment to testing ASD hypotheses is the lack of human cell models. Here, we reprogrammed fibroblasts to generate induced pluripotent stem cells, neural progenitor cells (NPCs) and neurons from ASD individuals with early brain overgrowth and non-ASD controls with normal brain size. ASD-derived NPCs display increased cell proliferation because of dysregulation of a β-catenin/BRN2 transcriptional cascade. ASD-derived neurons display abnormal neurogenesis and reduced synaptogenesis leading to functional defects in neuronal networks. Interestingly, defects in neuronal networks could be rescued by insulin growth factor 1 (IGF-1), a drug that is currently in clinical trials for ASD. This work demonstrates that selection of ASD subjects based on endophenotypes unraveled biologically relevant pathway disruption and revealed a potential cellular mechanism for the therapeutic effect of IGF-1.
Collapse
Affiliation(s)
| | - Haim Belinson
- University of California San Francisco, Department of Pediatrics, Institute for Human Genetics, CA 94143, USA
| | - Yuan Tian
- University of California Los Angeles, Program in Neurogenetics, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, Los Angeles, CA 90402, USA
| | - Beatriz C. Freitas
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| | - Chen Fu
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, OH 44106, USA
| | | | - Patricia Beltrao-Braga
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
- University of São Paulo, Department of Obstetrics, Department of Surgery, Center for Cellular and Molecular Therapy, São Paulo, Brazil
| | - Cleber A. Trujillo
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| | - Ana P.D. Mendes
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Krishnan Padmanabhan
- University of Rochester School of Medicine and Dentistry, Department of Neuroscience, 601 Elmwood Avenue, Box 603 Rochester, NY 14642
| | - Yanelli Nunez
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| | - Jing Ou
- University of California Los Angeles, Program in Neurogenetics, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, Los Angeles, CA 90402, USA
| | - Himanish Ghosh
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Rebecca Wright
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Kristen Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Karen Pierce
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Lawrence Eichenfield
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Tiziano Pramparo
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Lisa Eyler
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Cynthia C. Barnes
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Eric Courchesne
- University of California San Diego, Department of Neurosciences, La Jolla, CA 92093, USA
| | - Daniel H. Geschwind
- University of California Los Angeles, Program in Neurogenetics, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, Los Angeles, CA 90402, USA
| | - Fred H. Gage
- The Salk Institute, Laboratory of Genetics, La Jolla, CA 92037, USA
| | - Anthony Wynshaw-Boris
- University of California San Francisco, Department of Pediatrics, Institute for Human Genetics, CA 94143, USA
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, OH 44106, USA
| | - Alysson R. Muotri
- University of California San Diego, Department of Pediatrics/Rady Children’s Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093-0695, USA
| |
Collapse
|
38
|
Durán-Carabali LE, Arcego DM, Odorcyk FK, Reichert L, Cordeiro JL, Sanches EF, Freitas LD, Dalmaz C, Pagnussat A, Netto CA. Prenatal and Early Postnatal Environmental Enrichment Reduce Acute Cell Death and Prevent Neurodevelopment and Memory Impairments in Rats Submitted to Neonatal Hypoxia Ischemia. Mol Neurobiol 2017; 55:3627-3641. [DOI: 10.1007/s12035-017-0604-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/08/2017] [Indexed: 12/11/2022]
|
39
|
Brown SM, Peters R, Lawrence AB. Up-regulation of IGF-1 in the frontal cortex of piglets exposed to an environmentally enriched arena. Physiol Behav 2017; 173:285-292. [PMID: 28238777 PMCID: PMC5358774 DOI: 10.1016/j.physbeh.2017.02.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/22/2017] [Accepted: 02/22/2017] [Indexed: 01/09/2023]
Abstract
Environmental enrichment (EE) is widely used in the life sciences to study effects of environment on the brain. In pigs, despite lack of EE being a key welfare issue there is little understanding of brain effects of EE in pigs. This project aimed to study the effects of exposure to an EE arena on piglet behaviours and on brain gene expression levels with a focus on IGF-1 and related genes. Eight litters of large white×landrace×Hampshire piglets were farrowed and raised in a free farrowing system (PigSAFE). At 42days of age, 6pigletsperlitter were given access to an enriched arena with plentiful peat, straw and space, (in groups of 4 made up of stable pairs) for 15min per day on 5 consecutive days to allow them to habituate to the apparatus. Piglet behaviours were recorded in the arena for 15min periods on 3 consecutive days. On the final day only one pair of test piglets per litter was given access to the arena. Brain tissue was collected within 45min of the test from piglets exposed to the arena on the day and their non-exposed littermate controls. RNA was extracted from the frontal cortex and QRT-PCR for selected genes run on a Stratgene MX3005P. In both the home pen and the EE arena litters spent the largest proportion of time engaging in foraging behaviour which was significantly increased in the enriched arena (t7=5.35, df=6, p=0.001). There were decreases in non-running play (t7=4.82, p=0.002) and inactivity (t7=4.6, p=0.002) in the arena. A significant fold change increase (FC=1.07, t=4.42, p=0.002) was observed in IGF-1 gene expression in the frontal cortex of piglets exposed to the enriched arena compared to those not exposed on the day of culling. No change in expression was observed in CSF1, the IGF-1 receptor gene nor in any of the binding proteins tested (IGFBP1-6). There was a weak tendency for increased expression of the neurotrophic factor BDNF1 (fold change: 1.03; t7=1.54, p=0.1). We believe this work is the first to explore effects of EE on pig brain physiology and development, and also points to a potential role for IGF-1 in brain effects of EE.
Collapse
Affiliation(s)
- Sarah M. Brown
- University of Edinburgh, Roslin Institute, Penicuik EH25 9RG, United Kingdom,Corresponding author.
| | - Rebecca Peters
- SRUC, West Mains Road, Edinburgh EH9 3JG, United Kingdom
| | | |
Collapse
|
40
|
Baroncelli L, Cenni MC, Melani R, Deidda G, Landi S, Narducci R, Cancedda L, Maffei L, Berardi N. Early IGF-1 primes visual cortex maturation and accelerates developmental switch between NKCC1 and KCC2 chloride transporters in enriched animals. Neuropharmacology 2017; 113:167-177. [PMID: 26924708 DOI: 10.1016/j.neuropharm.2016.02.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/01/2016] [Accepted: 02/24/2016] [Indexed: 12/17/2022]
Abstract
Environmental enrichment (EE) has a remarkable impact on brain development. Continuous exposure to EE from birth determines a significant acceleration of visual system maturation both at retinal and cortical levels. A pre-weaning enriched experience is sufficient to trigger the accelerated maturation of the visual system, suggesting that factors affected by EE during the first days of life might prime visual circuits towards a faster development. The search for such factors is crucial not only to gain a better understanding of the molecular hierarchy of brain development but also to identify molecular pathways amenable to be targeted to correct atypical brain developmental trajectories. Here, we showed that IGF-1 levels are increased in the visual cortex of EE rats as early as P6 and this is a crucial event for setting in motion the developmental program induced by EE. Early intracerebroventricular (i.c.v.) infusion of IGF-1 in standard rats was sufficient to mimic the action of EE on visual acuity development, whereas blocking IGF-1 signaling by i.c.v. injections of the IGF-1 receptor antagonist JB1 prevented the deployment of EE effects. Early IGF-1 decreased the ratio between the expression of NKCC1 and KCC2 cation/chloride transporters, and the reversal potential for GABAAR-driven Cl- currents (ECl) was shifted toward more negative potentials, indicating that IGF-1 is a crucial factor in accelerating the maturation of GABAergic neurotransmission and promoting the developmental switch of GABA polarity from excitation to inhibition. In addition, early IGF-1 promoted a later occurring increase in its own expression, suggesting a priming effect of early IGF-1 in driving post-weaning cortical maturation.
Collapse
Affiliation(s)
- Laura Baroncelli
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy.
| | | | - Riccardo Melani
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy; Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, Piazza San Marco 4, I-50121, Florence, Italy
| | - Gabriele Deidda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genoa, Italy
| | - Silvia Landi
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy
| | - Roberta Narducci
- Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, Piazza San Marco 4, I-50121, Florence, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genoa, Italy
| | - Lamberto Maffei
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy; Laboratory of Neurobiology, Scuola Normale Superiore, Piazza Cavalieri 7, I-56126, Pisa, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy; Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, Piazza San Marco 4, I-50121, Florence, Italy
| |
Collapse
|
41
|
Wrigley S, Arafa D, Tropea D. Insulin-Like Growth Factor 1: At the Crossroads of Brain Development and Aging. Front Cell Neurosci 2017; 11:14. [PMID: 28203146 PMCID: PMC5285390 DOI: 10.3389/fncel.2017.00014] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/16/2017] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor 1 (IGF1) is a polypeptide hormone structurally similar to insulin. It is central to the somatotropic axis, acting downstream of growth hormone (GH). It activates both the mitogen-activated protein (MAP) kinase and PI3K signaling pathways, acting in almost every tissue in the body to promote tissue growth and maturation through upregulation of anabolic processes. Overall GH and IGF1 signaling falls with age, suggesting that it is this reduced IGF1 activity that leads to age-related changes in organisms. However, mutations that reduce IGF1-signaling activity can dramatically extend the lifespan of organisms. Therefore, the role of IGF1 in the overall aging process is unclear. This review article will focus on the role of IGF1 in brain development and aging. The evidence points towards a role for IGF1 in neurodevelopment both prenatally and in the early post-natal period, and in plasticity and remodeling throughout life. This review article will then discuss the hallmarks of aging and cognitive decline associated with falls in IGF1 levels towards the end of life. Finally, the role of IGF1 will be discussed within the context of both neuropsychiatric disorders caused by impaired development of the nervous system, and neurodegenerative disorders associated with aging. IGF1 and its derivatives are shown to improve the symptoms of certain neuropsychiatric disorders caused by deranged neurodevelopment and these effects have been correlated with changes in the underlying biology in both in vitro and in vivo studies. On the other hand, studies looking at IGF1 in neurodegenerative diseases have been conflicting, supporting both a role for increased and decreased IGF1 signaling in the underlying pathogenesis of these diseases.
Collapse
Affiliation(s)
- Sarah Wrigley
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Donia Arafa
- School of Medicine, Trinity College Dublin Dublin, Ireland
| | - Daniela Tropea
- Neuropsychiatric Genetics, Trinity Translational Medicine Institute St. James HospitalDublin, Ireland; Institute of Neuroscience, Trinity College DublinDublin, Ireland
| |
Collapse
|
42
|
Dai H, Goto YI, Itoh M. Insulin-Like Growth Factor Binding Protein-3 Deficiency Leads to Behavior Impairment with Monoaminergic and Synaptic Dysfunction. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:390-400. [PMID: 28088287 DOI: 10.1016/j.ajpath.2016.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/22/2016] [Accepted: 10/05/2016] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factor binding protein (IGFBP)-3 regulates IGF bioactivity, induces apoptosis, and inhibits cell growth independent of IGFs, but the functional role of IGFBP3 in the brain is not clear. In the present study, we revealed the effect of IGFBP3 on the brain by characterizing the phenotype of Igfbp3-null mice. Compared with wild-type mice, Igfbp3-null mice had significantly decreased IGF-1 content in the brain but no change in weights of brain and body. In Igfbp3-null mice, the number of dendritic spines was significantly reduced, and the dendritic diameter was thickening. In addition, in Igfbp3-null mice, a decrease in phosphorylated Akt and ERK1/2 significantly reduced PSD-95 expression, and GAD65/67 expression was significantly decreased. These results indicate that IGFBP3 deficiency impairs neuronal structure and signaling. In behavioral studies, Igfbp3-null mice were hyperactive, and a Y-maze alternation test revealed impaired spatial working memory but no anxiety-like behavior. Monoaminergic analysis using high-performance liquid chromatography indicated that Igfbp3-null mice had lower levels of dopamine and serotonin compared with wild-type mice, suggesting an abnormal monoaminergic neurotransmission. In conclusion, our studies found that the deletion of IGFBP3 results in behavioral impairments that are associated with abnormal synaptic function and monoaminergic neurotransmission, which helps to characterize the critical role of IGFBP3 in the brain.
Collapse
Affiliation(s)
- Hongmei Dai
- Department of Mental Retardation and Birth Defect Research, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yu-Ichi Goto
- Department of Mental Retardation and Birth Defect Research, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Masayuki Itoh
- Department of Mental Retardation and Birth Defect Research, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| |
Collapse
|
43
|
Bryan MR, Bowman AB. Manganese and the Insulin-IGF Signaling Network in Huntington's Disease and Other Neurodegenerative Disorders. ADVANCES IN NEUROBIOLOGY 2017; 18:113-142. [PMID: 28889265 PMCID: PMC6559248 DOI: 10.1007/978-3-319-60189-2_6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease resulting in motor impairment and death in patients. Recently, several studies have demonstrated insulin or insulin-like growth factor (IGF) treatment in models of HD, resulting in potent amelioration of HD phenotypes via modulation of the PI3K/AKT/mTOR pathways. Administration of IGF and insulin can rescue microtubule transport, metabolic function, and autophagy defects, resulting in clearance of Huntingtin (HTT) aggregates, restoration of mitochondrial function, amelioration of motor abnormalities, and enhanced survival. Manganese (Mn) is an essential metal to all biological systems but, in excess, can be toxic. Interestingly, several studies have revealed the insulin-mimetic effects of Mn-demonstrating Mn can activate several of the same metabolic kinases and increase peripheral and neuronal insulin and IGF-1 levels in rodent models. Separate studies have shown mouse and human striatal neuroprogenitor cell (NPC) models exhibit a deficit in cellular Mn uptake, indicative of a Mn deficiency. Furthermore, evidence from the literature reveals a striking overlap between cellular consequences of Mn deficiency (i.e., impaired function of Mn-dependent enzymes) and known HD endophenotypes including excitotoxicity, increased reactive oxygen species (ROS) accumulation, and decreased mitochondrial function. Here we review published evidence supporting a hypothesis that (1) the potent effect of IGF or insulin treatment on HD models, (2) the insulin-mimetic effects of Mn, and (3) the newly discovered Mn-dependent perturbations in HD may all be functionally related. Together, this review will present the intriguing possibility that intricate regulatory cross-talk exists between Mn biology and/or toxicology and the insulin/IGF signaling pathways which may be deeply connected to HD pathology and, perhaps, other neurodegenerative diseases (NDDs) and other neuropathological conditions.
Collapse
Affiliation(s)
- Miles R Bryan
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Center in Molecular Toxicology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
44
|
Dyer AH, Vahdatpour C, Sanfeliu A, Tropea D. The role of Insulin-Like Growth Factor 1 (IGF-1) in brain development, maturation and neuroplasticity. Neuroscience 2016; 325:89-99. [DOI: 10.1016/j.neuroscience.2016.03.056] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/29/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
|
45
|
Yu JH, Kim M, Seo JH, Cho SR. Brain Plasticity and Neurorestoration by Environmental Enrichment. BRAIN & NEUROREHABILITATION 2016. [DOI: 10.12786/bn.2016.9.e2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ji Hea Yu
- Department of Rehabilitation Medicine and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Yonsei Stem Cell Center, Avison Biomedical Research Center, Seoul, Korea
| | - MinGi Kim
- Department of Rehabilitation Medicine and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Jung Hwa Seo
- Department of Rehabilitation Medicine and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Sung-Rae Cho
- Department of Rehabilitation Medicine and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Yonsei Stem Cell Center, Avison Biomedical Research Center, Seoul, Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Begenisic T, Sansevero G, Baroncelli L, Cioni G, Sale A. Early environmental therapy rescues brain development in a mouse model of Down syndrome. Neurobiol Dis 2015; 82:409-419. [PMID: 26244989 DOI: 10.1016/j.nbd.2015.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/26/2015] [Accepted: 07/31/2015] [Indexed: 01/15/2023] Open
Abstract
Down syndrome (DS), the most common genetic disorder associated with intellectual disabilities, is an untreatable condition characterized by a number of developmental defects and permanent deficits in the adulthood. Ts65Dn mice, the major animal model for DS, display severe cognitive and synaptic plasticity defects closely resembling the human phenotype. Here, we employed a multidisciplinary approach to investigate, for the first time in developing Ts65Dn mice, the effects elicited by early environmental enrichment (EE) on brain maturation and function. We report that exposure to EE resulted in a robust increase in maternal care levels displayed by Ts65Dn mothers and led to a normalization of declarative memory abilities and hippocampal plasticity in trisomic offspring. The positive effects of EE on Ts65Dn phenotype were not limited to the cognitive domain, but also included a rescue of visual system maturation. The beneficial EE effects were accompanied by increased BDNF and correction of over-expression of the GABA vesicular transporter vGAT. These findings highlight the beneficial impact of early environmental stimuli and their potential for application in the treatment of major functional deficits in children with DS.
Collapse
Affiliation(s)
| | | | | | - Giovanni Cioni
- Department of Developmental Neuroscience, IRCCS Stella Maris, University of Pisa, Calambrone, I-56100 Pisa, Italy
| | | |
Collapse
|
47
|
Maturana-Teixeira S, Braga LEG, Carpi Santos R, Calaza KDC, Giestal-de-Araujo E, Leão-Ferreira LR. The (Na(+)/K (+))-ATPase activity in the developing rat retina: the role of insulin-like growth factor-I (IGF-I). Cell Mol Neurobiol 2015; 35:243-54. [PMID: 25274047 PMCID: PMC11486223 DOI: 10.1007/s10571-014-0119-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/23/2014] [Indexed: 02/04/2023]
Abstract
In this work, the (Na(+)/K(+))-ATPase activity was evaluated during the early stages of the postnatal development of rat retina and showed an almost three-time increase from P0 to P14. Expression of the three catalytic subunit isoforms (α1, α2, and α3) of the (Na(+)/K(+))-ATPase was also evaluated by immunoblot in the same period, but no correlation to the catalytic activity increment was observed. On the other hand, immunolocalization of these three α-catalytic isoforms in the developing retina showed an age-related pattern. Involvement of IGF-I in the stimulation of the (Na(+)/K(+))-ATPase was investigated. Our results demonstrate that the exogenous IGF-I (10 ng/mL) stimulates enzyme activity at the age of P7 only. Incubation of retinas with 10 μM I-OMe-AG 538 (inhibitor of the IGF-I receptor) indicates that the basal (Na(+)/K(+))-ATPase activity is sustained by endogenous IGF-I in P7 animals. These data were corroborated by an age-dependent decrease in the immunodetection of endogenous IGF-I as well as in the phosphorylation level of its cognate receptor in rat retina homogenates. The signaling pathway involved in IGF-I-induced modulation of the (Na(+)/K(+))-ATPase was also investigated. Our data show that the inhibitory effects induced by I-OMe-AG 538 and the PI 3-kinase inhibitor Ly 294002 on the basal (Na(+)/K(+))-ATPase activity were non-cumulative. Furthermore, IGF-I induced phosphorylation of PKB in a Ly 294002-sensitive manner. Together, these data demonstrate that the PI 3-kinase/PKB signaling pathway is involved in the IGF-I-sustained basal (Na(+)/K(+))-ATPase activity during the first 7 days of the postnatal development of rat retina.
Collapse
Affiliation(s)
- Sheila Maturana-Teixeira
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio De Janeiro CEP 24020-140 Brazil
| | - Luis Eduardo Gomes Braga
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio De Janeiro CEP 24020-140 Brazil
| | - Raul Carpi Santos
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio De Janeiro CEP 24020-140 Brazil
| | - Karin da Costa Calaza
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio De Janeiro CEP 24020-140 Brazil
| | - Elizabeth Giestal-de-Araujo
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio De Janeiro CEP 24020-140 Brazil
| | - Luiz Roberto Leão-Ferreira
- Programa de Neurociências, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio De Janeiro CEP 24020-140 Brazil
| |
Collapse
|
48
|
Cortical plasticity induced by transplantation of embryonic somatostatin or parvalbumin interneurons. Proc Natl Acad Sci U S A 2014; 111:18339-44. [PMID: 25489113 DOI: 10.1073/pnas.1421844112] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
GABAergic inhibition has been shown to play an important role in the opening of critical periods of brain plasticity. We recently have shown that transplantation of GABAergic precursors from the embryonic medial ganglionic eminence (MGE), the source of neocortical parvalbumin- (PV(+)) and somatostatin-expressing (SST(+)) interneurons, can induce a new period of ocular dominance plasticity (ODP) after the endogenous period has closed. Among the diverse subtypes of GABAergic interneurons PV(+) cells have been thought to play the crucial role in ODP. Here we have used MGE transplantation carrying a conditional allele of diphtheria toxin alpha subunit and cell-specific expression of Cre recombinase to deplete PV(+) or SST(+) interneurons selectively and to investigate the contributions of each of these types of interneurons to ODP. As expected, robust plasticity was observed in transplants containing PV(+) cells but in which the majority of SST(+) interneurons were depleted. Surprisingly, transplants in which the majority of PV(+) cells were depleted induced plasticity as effectively as those containing PV(+) cells. In contrast, depleting both cell types blocked induction of plasticity. These findings reveal that PV(+) cells do not play an exclusive role in ODP; SST(+) interneurons also can drive cortical plasticity and contribute to the reshaping of neural networks. The ability of both PV(+) and SST(+) interneurons to induce de novo cortical plasticity could help develop new therapeutic approaches for brain repair.
Collapse
|
49
|
Deidda G, Allegra M, Cerri C, Naskar S, Bony G, Zunino G, Bozzi Y, Caleo M, Cancedda L. Early depolarizing GABA controls critical-period plasticity in the rat visual cortex. Nat Neurosci 2014; 18:87-96. [PMID: 25485756 PMCID: PMC4338533 DOI: 10.1038/nn.3890] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/03/2014] [Indexed: 12/16/2022]
Abstract
Hyperpolarizing and inhibitory GABA regulates critical periods for plasticity in sensory cortices. Here we examine the role of early, depolarizing GABA in the control of plasticity mechanisms. We report that brief interference with depolarizing GABA during early development prolonged critical-period plasticity in visual cortical circuits without affecting the overall development of the visual system. The effects on plasticity were accompanied by dampened inhibitory neurotransmission, downregulation of brain-derived neurotrophic factor (BDNF) expression and reduced density of extracellular matrix perineuronal nets. Early interference with depolarizing GABA decreased perinatal BDNF signaling, and a pharmacological increase of BDNF signaling during GABA interference rescued the effects on plasticity and its regulators later in life. We conclude that depolarizing GABA exerts a long-lasting, selective modulation of plasticity of cortical circuits by a strong crosstalk with BDNF.
Collapse
Affiliation(s)
- Gabriele Deidda
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy
| | - Manuela Allegra
- 1] Scuola Normale Superiore, Pisa, Italy. [2] CNR Neuroscience Institute, Pisa, Italy
| | | | - Shovan Naskar
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy
| | - Guillaume Bony
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy
| | - Giulia Zunino
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Yuri Bozzi
- 1] CNR Neuroscience Institute, Pisa, Italy. [2] Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | | | - Laura Cancedda
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
50
|
Transformation of cortical and hippocampal neural circuit by environmental enrichment. Neuroscience 2014; 280:282-98. [PMID: 25242640 DOI: 10.1016/j.neuroscience.2014.09.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/10/2014] [Accepted: 09/10/2014] [Indexed: 12/17/2022]
Abstract
It has been half a century since brain volume enlargement was first reported in animals reared in an enriched environment (EE). As EE animals show improved memory task performance, exposure to EE has been a useful model system for studying the effects of experience on brain plasticity. We review EE-induced neural changes in the cerebral cortex and hippocampus focusing mainly on works published in the recent decade. The review is organized in three large domains of changes: anatomical, electrophysiological, and molecular changes. Finally, we discuss open issues and future outlook toward better understanding of EE-induced neural changes.
Collapse
|