1
|
Wu R, Xiong Y, Gu Y, Cao LY, Zhang SY, Song ZX, Fan P, Lin L. Traditional Pediatric Massage Enhanced Hippocampal GR, BDNF and IGF-1 Expressions and Exerted an Anti-depressant Effect in an Adolescent Rat Model of CUMS-induced Depression. Neuroscience 2024; 542:47-58. [PMID: 38364964 DOI: 10.1016/j.neuroscience.2024.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/18/2024]
Abstract
This study aimed to investigate the anti-depressant effect of traditional pediatric massage (TPM) in adolescent rats and its possible mechanism. The adolescent depression model in rats was established by using chronic unpredictable mild stress (CUMS). All rats were randomly divided into five groups (seven per group), including the groups of control (CON), CUMS, CUMS with TPM, CUMS with back stroking massage (BSM) and CUMS with fluoxetine (FLX). The tests of sucrose preference, Morris water maze and elevated plus maze were used to evaluate depression-related behaviors. Plasma corticosterone (CORT) level was measured by ELISA. The gene and protein expressions of glucocorticoid receptor (GR), brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1 (IGF-1) were measured by RT-qPCR and IHC respectively. The results showed that CUMS induced depression-related behaviors in the adolescent rats, along with decreased weight gain and reduced hippocampal expressions of GR, IGF-1 and BDNF. TPM could effectively prevent depression-related behaviors in CUMS-exposed adolescent rats, manifested as increasing weight gain, sucrose consumption, ratio of open-arm entry, times of crossing the specific quadrant and shortening escape latency. TPM also decreased CORT level in plasma, together with enhancing expressions of GR, IGF-1 and BDNF in the hippocampus. These results may support the clinical application of TPM to prevent and treat adolescent depression.
Collapse
Affiliation(s)
- Rong Wu
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Xiong
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yun Gu
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, China
| | - Li-Yue Cao
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shu-Ying Zhang
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi-Xiu Song
- College of Health and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pu Fan
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lin Lin
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Han Y, Yuan M, Guo YS, Shen XY, Gao ZK, Bi X. The role of enriched environment in neural development and repair. Front Cell Neurosci 2022; 16:890666. [PMID: 35936498 PMCID: PMC9350910 DOI: 10.3389/fncel.2022.890666] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/29/2022] [Indexed: 12/01/2022] Open
Abstract
In addition to genetic information, environmental factors play an important role in the structure and function of nervous system and the occurrence and development of some nervous system diseases. Enriched environment (EE) can not only promote normal neural development through enhancing neuroplasticity but also play a nerve repair role in restoring functional activities during CNS injury by morphological and cellular and molecular adaptations in the brain. Different stages of development after birth respond to the environment to varying degrees. Therefore, we systematically review the pro-developmental and anti-stress value of EE during pregnancy, pre-weaning, and “adolescence” and analyze the difference in the effects of EE and its sub-components, especially with physical exercise. In our exploration of potential mechanisms that promote neurodevelopment, we have found that not all sub-components exert maximum value throughout the developmental phase, such as animals that do not respond to physical activity before weaning, and that EE is not superior to its sub-components in all respects. EE affects the developing and adult brain, resulting in some neuroplastic changes in the microscopic and macroscopic anatomy, finally contributing to enhanced learning and memory capacity. These positive promoting influences are particularly prominent regarding neural repair after neurobiological disorders. Taking cerebral ischemia as an example, we analyzed the molecular mediators of EE promoting repair from various dimensions. We found that EE does not always lead to positive effects on nerve repair, such as infarct size. In view of the classic issues such as standardization and relativity of EE have been thoroughly discussed, we finally focus on analyzing the essentiality of the time window of EE action and clinical translation in order to devote to the future research direction of EE and rapid and reasonable clinical application.
Collapse
Affiliation(s)
- Yu Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Mei Yuan
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yi-Sha Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Graduate School, Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen-Kun Gao
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Graduate School, Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Xia Bi
| |
Collapse
|
3
|
Knipper M, Singer W, Schwabe K, Hagberg GE, Li Hegner Y, Rüttiger L, Braun C, Land R. Disturbed Balance of Inhibitory Signaling Links Hearing Loss and Cognition. Front Neural Circuits 2022; 15:785603. [PMID: 35069123 PMCID: PMC8770933 DOI: 10.3389/fncir.2021.785603] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Neuronal hyperexcitability in the central auditory pathway linked to reduced inhibitory activity is associated with numerous forms of hearing loss, including noise damage, age-dependent hearing loss, and deafness, as well as tinnitus or auditory processing deficits in autism spectrum disorder (ASD). In most cases, the reduced central inhibitory activity and the accompanying hyperexcitability are interpreted as an active compensatory response to the absence of synaptic activity, linked to increased central neural gain control (increased output activity relative to reduced input). We here suggest that hyperexcitability also could be related to an immaturity or impairment of tonic inhibitory strength that typically develops in an activity-dependent process in the ascending auditory pathway with auditory experience. In these cases, high-SR auditory nerve fibers, which are critical for the shortest latencies and lowest sound thresholds, may have either not matured (possibly in congenital deafness or autism) or are dysfunctional (possibly after sudden, stressful auditory trauma or age-dependent hearing loss linked with cognitive decline). Fast auditory processing deficits can occur despite maintained basal hearing. In that case, tonic inhibitory strength is reduced in ascending auditory nuclei, and fast inhibitory parvalbumin positive interneuron (PV-IN) dendrites are diminished in auditory and frontal brain regions. This leads to deficits in central neural gain control linked to hippocampal LTP/LTD deficiencies, cognitive deficits, and unbalanced extra-hypothalamic stress control. Under these conditions, a diminished inhibitory strength may weaken local neuronal coupling to homeostatic vascular responses required for the metabolic support of auditory adjustment processes. We emphasize the need to distinguish these two states of excitatory/inhibitory imbalance in hearing disorders: (i) Under conditions of preserved fast auditory processing and sustained tonic inhibitory strength, an excitatory/inhibitory imbalance following auditory deprivation can maintain precise hearing through a memory linked, transient disinhibition that leads to enhanced spiking fidelity (central neural gain⇑) (ii) Under conditions of critically diminished fast auditory processing and reduced tonic inhibitory strength, hyperexcitability can be part of an increased synchronization over a broader frequency range, linked to reduced spiking reliability (central neural gain⇓). This latter stage mutually reinforces diminished metabolic support for auditory adjustment processes, increasing the risks for canonical dementia syndromes.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- *Correspondence: Marlies Knipper,
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Kerstin Schwabe
- Experimental Neurosurgery, Department of Neurosurgery, Hannover Medical School, Hanover, Germany
| | - Gisela E. Hagberg
- Department of Biomedical Magnetic Resonance, University Hospital Tübingen (UKT), Tübingen, Germany
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Yiwen Li Hegner
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Christoph Braun
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Rüdiger Land
- Department of Experimental Otology, Institute for Audioneurotechnology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
4
|
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: 4] [Impact Index Per Article: 1.3] [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
|
5
|
Peerboom C, Wierenga CJ. The postnatal GABA shift: A developmental perspective. Neurosci Biobehav Rev 2021; 124:179-192. [PMID: 33549742 DOI: 10.1016/j.neubiorev.2021.01.024] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/13/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
GABA is the major inhibitory neurotransmitter that counterbalances excitation in the mature brain. The inhibitory action of GABA relies on the inflow of chloride ions (Cl-), which hyperpolarizes the neuron. In early development, GABA signaling induces outward Cl- currents and is depolarizing. The postnatal shift from depolarizing to hyperpolarizing GABA is a pivotal event in brain development and its timing affects brain function throughout life. Altered timing of the postnatal GABA shift is associated with several neurodevelopmental disorders. Here, we argue that the postnatal shift from depolarizing to hyperpolarizing GABA represents the final shift in a sequence of GABA shifts, regulating proliferation, migration, differentiation, and finally plasticity of developing neurons. Each developmental GABA shift ensures that the instructive role of GABA matches the circumstances of the developing network. Sensory input may be a crucial factor in determining proper timing of the postnatal GABA shift. A developmental perspective is necessary to interpret the full consequences of a mismatch between connectivity, activity and GABA signaling during brain development.
Collapse
Affiliation(s)
- Carlijn Peerboom
- Cell Biology, Neurobiology and Biophysics, Biology Department, Faculty of Science, Utrecht University, 3584 CH, Utrecht, the Netherlands
| | - Corette J Wierenga
- Cell Biology, Neurobiology and Biophysics, Biology Department, Faculty of Science, Utrecht University, 3584 CH, Utrecht, the Netherlands.
| |
Collapse
|
6
|
Gao L, Kita T, Katsuno T, Yamamoto N, Omori K, Nakagawa T. Insulin-Like Growth Factor 1 on the Maintenance of Ribbon Synapses in Mouse Cochlear Explant Cultures. Front Cell Neurosci 2020; 14:571155. [PMID: 33132846 PMCID: PMC7579230 DOI: 10.3389/fncel.2020.571155] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/27/2020] [Indexed: 01/31/2023] Open
Abstract
Hearing loss has become one of the most common disabilities worldwide. The synaptic connections between inner hair cells (IHCs) and spiral ganglion neurons have specialized synaptic constructions, termed ribbon synapses, which are important for auditory function. The ribbon synapses in the cochlea are quite vulnerable to various insults. As such, the maintenance of ribbon synapses is important for ensuring hearing function. Insulin-like growth factor 1 (IGF1) plays a critical role in the development and maintenance of the cochlea and has the potential to protect cochlear hair cells from various insults. In this study, we examined the role of IGF1 in the maintenance of ribbon synapses in cochlear explants of postnatal day four mice. We cultured cochlear explants with an IGF1 receptor antagonist, JB1, which is an IGF1 peptide analog. Results showed that exposure to JB1 for 24 h resulted in the loss of ribbon synapses. After an additional 24-h culture without JB1, the number of ribbon synapses spontaneously recovered. The application of exogenous IGF1 showed two different aspects of ribbon synapses. Low doses of exogenous IGF1 promoted the recovery of ribbon synapses, while it compromised the spontaneous recovery of ribbon synapses at high doses. Altogether, these results indicate that the paracrine or autocrine release of IGF1 in the cochlea plays a crucial role in the maintenance of cochlear ribbon synapses.
Collapse
Affiliation(s)
- Li Gao
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Kita
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuya Katsuno
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
7
|
Auer T, Schreppel P, Erker T, Schwarzer C. Impaired chloride homeostasis in epilepsy: Molecular basis, impact on treatment, and current treatment approaches. Pharmacol Ther 2020; 205:107422. [DOI: 10.1016/j.pharmthera.2019.107422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
|
8
|
González Fleitas MF, Devouassoux JD, Aranda ML, Calanni JS, Chianelli MS, Dorfman D, Rosenstein RE. Enriched environment provides neuroprotection against experimental glaucoma. J Neurochem 2019; 152:103-121. [PMID: 31587281 DOI: 10.1111/jnc.14885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/13/2019] [Accepted: 10/01/2019] [Indexed: 12/25/2022]
Abstract
Glaucoma is one of the most frequent causes of visual impairment worldwide, and involves selective damage to retinal ganglion cells (RGCs) and their axons. We analyzed the effect of enriched environment (EE) housing on the optic nerve, and retinal alterations in an induced model of ocular hypertension. For this purpose, male Wistar rats were weekly injected with vehicle or chondroitin sulfate (CS) into the eye anterior chamber for 10 weeks and housed in standard environment or EE. EE housing prevented the effect of experimental glaucoma on visual evoked potentials, retinal anterograde transport, phosphorylated neurofilament-immunoreactivity, axon number, microglial/macrophage reactivity (ionized calcium binding adaptor molecule 1-immunoreactivity), and astrocytosis (glial fibrillary acidic protein-immunostaining), as well as oligodendrocytes alterations (luxol fast blue staining, and myelin basic protein-immunoreactivity) in the proximal portion of the optic nerve. Moreover EE prevented the increase in ionized calcium binding adaptor molecule-1 levels, and RGC loss (Brn3a-immunoreactivity) in the retina from hypertensive eyes. EE increased retinal brain-derived neurotrophic factor levels. When EE housing started after 6 weeks of ocular hypertension, a preservation of visual evoked potentials amplitude, axon, and Brn3a(+) RGC number was observed. Taken together, these results suggest that EE preserved visual functions, reduced optic nerve axoglial alterations, and protected RGCs against glaucomatous damage.
Collapse
Affiliation(s)
- María F González Fleitas
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Julián D Devouassoux
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Marcos L Aranda
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Juan S Calanni
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Monica S Chianelli
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Damián Dorfman
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Ruth E Rosenstein
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| |
Collapse
|
9
|
Turkson S, Kloster A, Hamilton PJ, Neigh GN. Neuroendocrine drivers of risk and resilience: The influence of metabolism & mitochondria. Front Neuroendocrinol 2019; 54:100770. [PMID: 31288042 PMCID: PMC6886586 DOI: 10.1016/j.yfrne.2019.100770] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/20/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
The manifestation of risk versus resilience has been considered from varying perspectives including genetics, epigenetics, early life experiences, and type and intensity of the challenge with which the organism is faced. Although all of these factors are central to determining risk and resilience, the current review focuses on what may be a final common pathway: metabolism. When an organism is faced with a perturbation to the environment, whether internal or external, appropriate energy allocation is essential to resolving the divergence from equilibrium. This review examines the potential role of metabolism in the manifestation of stress-induced neural compromise. In addition, this review details the current state of knowledge on neuroendocrine factors which are poised to set the tone of the metabolic response to a systemic challenge. The goal is to provide an essential framework for understanding stress in a metabolic context and appreciation for key neuroendocrine signals.
Collapse
Affiliation(s)
- Susie Turkson
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Alix Kloster
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Peter J Hamilton
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Gretchen N Neigh
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
10
|
Kentner AC, Cryan JF, Brummelte S. Resilience priming: Translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019; 61:350-375. [PMID: 30311210 PMCID: PMC6447439 DOI: 10.1002/dev.21775] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the increasing attention to early life adversity and its long-term consequences on health, behavior, and the etiology of neurodevelopmental disorders, our understanding of the adaptations and interventions that promote resiliency and rescue against such insults are underexplored. Specifically, investigations of the perinatal period often focus on negative events/outcomes. In contrast, positive experiences (i.e. enrichment/parental care//healthy nutrition) favorably influence development of the nervous and endocrine systems. Moreover, some stressors result in adaptations and demonstrations of later-life resiliency. This review explores the underlying mechanisms of neuroplasticity that follow some of these early life experiences and translates them into ideas for interventions in pediatric settings. The emerging role of the gut microbiome in mediating stress susceptibility is also discussed. Since many negative outcomes of early experiences are known, it is time to identify mechanisms and mediators that promote resiliency against them. These range from enrichment, quality parental care, dietary interventions and those that target the gut microbiota.
Collapse
Affiliation(s)
- Amanda C. Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115,
| | - John F. Cryan
- Dept. Anatomy & Neuroscience & APC Microbiome Institute, University College Cork, College Rd., Cork, Ireland,
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202,
| |
Collapse
|
11
|
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: 0.8] [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
|
12
|
Schulte JT, Wierenga CJ, Bruining H. Chloride transporters and GABA polarity in developmental, neurological and psychiatric conditions. Neurosci Biobehav Rev 2018; 90:260-271. [PMID: 29729285 DOI: 10.1016/j.neubiorev.2018.05.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
Neuronal chloride regulation is a determinant factor for the dynamic tuning of GABAergic inhibition during and beyond brain development. This regulation is mainly dependent on the two co-transporters K+/Cl- co-transporter KCC2 and Na+/K+/Cl- co-transporter NKCC1, whose activity can decrease or increase neuronal chloride concentrations respectively. Altered expression and/or activity of either of these co-transporters has been associated with a wide variety of brain disorders including developmental disorders, epilepsy, schizophrenia and stroke. Here, we review current knowledge on chloride transporter expression and activity regulation and highlight the intriguing potential for existing and future interventions to support chloride homeostasis across a wide range of mental disorders and neurological conditions.
Collapse
Affiliation(s)
- Joran T Schulte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands
| | - Corette J Wierenga
- Division of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Hilgo Bruining
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands.
| |
Collapse
|
13
|
Tang X, Chen F, Lin Q, You Y, Ke J, Zhao S. Bone marrow mesenchymal stem cells repair the hippocampal neurons and increase the expression of IGF-1 after cardiac arrest in rats. Exp Ther Med 2017; 14:4312-4320. [PMID: 29067112 PMCID: PMC5647699 DOI: 10.3892/etm.2017.5059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 08/21/2017] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to investigate the beneficial effects and underlying mechanisms of bone marrow mesenchymal stem cells (BMSCs) on global ischemic hypoxic brain injury. Cells collected from the femurs and tibias of male Sprague Dawley rats were used to generate BMSCs following three culture passages. A rate model of cardiac arrest (CA) was induced by asphyxia. One hour following return of spontaneous circulation (ROSC), BMSCs were transplanted through injection into the tail vein. Neurological status was assessed using modified neurological severity score (mNSS) tests 1, 3 and 7 days following ROSC. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemical staining were used to detect insulin-like growth factor 1 (IGF-1) expression in the hippocampus. Furthermore, double-fluorescent labeling of green fluorescent protein (GFP) and IGF-1 was used to detect the IGF-1 expression in transplanted BMSCs. Serum levels of protein S100-B were examined using ELISA. GFP-labeled BMSCs were observed in the hippocampus at 1, 3 and 7 days post transplantation through fluorescent microscopy. BMSC transplantation resulted in reduced protein S100-B levels. The mNSS of the BMSC-treatment group was significantly reduced compared with that of the CA group. The RT-qPCR analysis and immunohistochemistry results demonstrated that BMSC treatment significantly increased IGF-1 expression in the hippocampus. In addition, the double-fluorescent labeling results demonstrated that transplanted BMSCs expressed IGF-1 in the hippocampus. The results of the present study suggest that BMSC treatment promotes the recovery of cerebral function following CA in rats possibly through the secretion of IGF-1.
Collapse
Affiliation(s)
- Xiahong Tang
- Department of Emergency, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350009, P.R. China.,Fujian Provincial Institute of Emergency Medicien, Fuzhou, Fujian 350009, P.R. China.,Fujian Emergency Medical Center, Fuzhou, Fujian 350009, P.R. China
| | - Feng Chen
- Department of Emergency, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350009, P.R. China.,Fujian Provincial Institute of Emergency Medicien, Fuzhou, Fujian 350009, P.R. China.,Fujian Emergency Medical Center, Fuzhou, Fujian 350009, P.R. China
| | - Qinming Lin
- Department of Emergency, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350009, P.R. China.,Fujian Provincial Institute of Emergency Medicien, Fuzhou, Fujian 350009, P.R. China.,Fujian Emergency Medical Center, Fuzhou, Fujian 350009, P.R. China
| | - Yan You
- Department of Emergency, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350009, P.R. China.,Fujian Provincial Institute of Emergency Medicien, Fuzhou, Fujian 350009, P.R. China.,Fujian Emergency Medical Center, Fuzhou, Fujian 350009, P.R. China
| | - Jun Ke
- Department of Emergency, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350009, P.R. China.,Fujian Provincial Institute of Emergency Medicien, Fuzhou, Fujian 350009, P.R. China.,Fujian Emergency Medical Center, Fuzhou, Fujian 350009, P.R. China
| | - Shen Zhao
- Department of Emergency, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350009, P.R. China.,Fujian Provincial Institute of Emergency Medicien, Fuzhou, Fujian 350009, P.R. China.,Fujian Emergency Medical Center, Fuzhou, Fujian 350009, P.R. China
| |
Collapse
|
14
|
Rosso G, Zanardini R, Chiodelli DF, Ferrari C, Gennarelli M, Bocchio-Chiavetto L. Serum Levels of Insulin-Like Growth Factor-1 and Obsessive-Compulsive Disorder: A Case-Control Study. Neuropsychobiology 2017; 74:15-21. [PMID: 27459640 DOI: 10.1159/000446918] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/18/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Recent findings suggest an involvement of insulin-like growth factor-1 (IGF-1) in the pathogenesis of many psychiatric disorders; however, there is a lack of data regarding IGF-1 in patients with obsessive-compulsive disorder (OCD). The aims of the present study were (1) to analyze putative alterations of IGF-1 serum content in patients with OCD compared to patients with major depressive disorder (MDD) and healthy controls, and (2) to analyze putative changes of IGF-1 levels during drug treatment in subjects with OCD compared to patients with MDD. METHODS We recruited 40 OCD patients, 37 MDD patients, and 43 healthy controls. All participants were adults. Serum IGF-1 levels were measured by the ELISA method on venous blood samples collected at baseline and after 10 ± 1 weeks of drug treatment. RESULTS IGF-1 levels were increased in OCD patients compared to controls (149.9 ± 60.2 vs. 121.2 ± 51.6 ng/ml; p = 0.040). No correlations were observed between baseline IGF-1 levels, clinical features, and response to treatment at follow-up in OCD or MDD patients. No changes in serum IGF-1 were observed after drug treatment. CONCLUSION Our results show for the first time that serum IGF-1 levels are altered in patients with OCD. Further research on the role of IGF-1 in OCD is warranted.
Collapse
Affiliation(s)
- Gianluca Rosso
- Psychiatric Unit, San Luigi Gonzaga Hospital of Orbassano, Neuroscience Department, University of Turin, Orbassano, IRCCS San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | | | | | | | | |
Collapse
|
15
|
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: 3.1] [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
|
16
|
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: 153] [Impact Index Per Article: 21.9] [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
|
17
|
Kong D, Gong L, Arnold E, Shanmugam S, Fort PE, Gardner TW, Abcouwer SF. Insulin-like growth factor 1 rescues R28 retinal neurons from apoptotic death through ERK-mediated BimEL phosphorylation independent of Akt. Exp Eye Res 2016; 151:82-95. [PMID: 27511131 DOI: 10.1016/j.exer.2016.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/27/2016] [Accepted: 08/05/2016] [Indexed: 10/21/2022]
Abstract
Insulin-like growth factor 1 (IGF-1) can provide long-term neurotrophic support by activation of Akt, inhibition of FoxO nuclear localization and suppression of Bim gene transcription in multiple neuronal systems. However, MEK/ERK activation can also promote neuron survival through phosphorylation of BimEL. We explored the contribution of the PI3K/Akt/FoxO and MEK/ERK/BimEL pathways in IGF-1 stimulated survival after serum deprivation (SD) of R28 cells differentiated to model retinal neurons. IGF-1 caused rapid activation of Akt leading to FoxO1/3-T32/T24 phosphorylation, and prevented FoxO1/3 nuclear translocation and Bim mRNA upregulation in response to SD. IGF-1 also caused MAPK/MEK pathway activation as indicated by ERK1/2-T202/Y204 and Bim-S65 phosphorylation. Overexpression of FoxO1 increased Bim mRNA expression and amplified the apoptotic response to SD without shifting the serum response curve. Inhibition of Akt activation with LY294002 or by Rictor knockdown did not block the protective effect of IGF-1, while inhibition of MEK activity with PD98059 prevented Bim phosphorylation and blocked IGF-1 protection. In addition, knockdown of Bim expression was protective during SD, while co-silencing of FoxO1 and Fox03 expression had little effect. Thus, the PI3K/Akt/FoxO pathway was not essential for protection from SD-induced apoptosis by IGF-1 in R28 cells. Instead, IGF-1 protection was dependent on activation of the MEK/ERK pathway leading to BimEL phosphorylation, which is known to prevent Bax/Bak oligomerization and activation of the intrinsic mitochondrial apoptosis pathway. These studies demonstrate the requirement of the MEK/ERK pathway in a model of retinal neuron cell survival and highlight the cell specificity for IGF-1 signaling in this response.
Collapse
Affiliation(s)
- Dejuan Kong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States
| | - Lijie Gong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States
| | - Edith Arnold
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States
| | - Sumathi Shanmugam
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States
| | - Patrice E Fort
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States
| | - Thomas W Gardner
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States
| | - Steven F Abcouwer
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States.
| |
Collapse
|
18
|
Abstract
UNLABELLED Tauopathies are neurodegenerative diseases characterized by intraneuronal inclusions of hyperphosphorylated tau protein and abnormal expression of brain-derived neurotrophic factor (BDNF), a key modulator of neuronal survival and function. The severity of both these pathological hallmarks correlate with the degree of cognitive impairment in patients. However, how tau pathology specifically modifies BDNF signaling and affects neuronal function during early prodromal stages of tauopathy remains unclear. Here, we report that the mild tauopathy developing in retinal ganglion cells (RGCs) of the P301S tau transgenic (P301S) mouse induces functional retinal changes by disrupting BDNF signaling via the TrkB receptor. In adult P301S mice, the physiological visual response of RGCs to pattern light stimuli and retinal acuity decline significantly. As a consequence, the activity-dependent secretion of BDNF in the vitreous is impaired in P301S mice. Further, in P301S retinas, TrkB receptors are selectively upregulated, but uncoupled from downstream extracellular signal-regulated kinase (ERK) 1/2 signaling. We also show that the impairment of TrkB signaling is triggered by tau pathology and mediates the tau-induced dysfunction of visual response. Overall our results identify a neurotrophin-mediated mechanism by which tau induces neuronal dysfunction during prodromal stages of tauopathy and define tau-driven pathophysiological changes of potential value to support early diagnosis and informed therapeutic decisions. SIGNIFICANCE STATEMENT This work highlights the potential molecular mechanisms by which initial tauopathy induces neuronal dysfunction. Combining clinically used electrophysiological techniques (i.e., electroretinography) and molecular analyses, this work shows that in a relevant model of early tauopathy, the retina of the P301S mutant human tau transgenic mouse, mild tau pathology results in functional changes of neuronal activity, likely due to selective impairment of brain-derived neurotrophic factor signaling via its receptor, TrkB. These findings may have important translational implications for early diagnosis in a subset of Alzheimer's disease patients with early visual symptoms and emphasize the need to clarify the pathophysiological changes associated with distinct tauopathy stages to support informed therapeutic decisions and guide drug discovery.
Collapse
|
19
|
Greifzu F, Kalogeraki E, Löwel S. Environmental enrichment preserved lifelong ocular dominance plasticity, but did not improve visual abilities. Neurobiol Aging 2016; 41:130-137. [PMID: 27103526 DOI: 10.1016/j.neurobiolaging.2016.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/01/2016] [Accepted: 02/11/2016] [Indexed: 11/28/2022]
Abstract
In standard cage (SC)-raised mice, ocular dominance (OD) plasticity of the primary visual cortex (V1) induced by monocular deprivation (MD) is maximal in juveniles, declines in adults, and is absent beyond postnatal day (PD) 110. Raising mice in an enriched environment (EE) preserved a juvenile-like OD plasticity after 7 days of MD until at least PD196, mediated by reductions of deprived eye responses in V1. Whether the sensitive phase for OD plasticity can be prolonged into older age and whether long-term EE modifies visual abilities was not yet known. Here, we demonstrate that EE raising enables lifelong OD plasticity. In contrast to PD200 EE-mice, the preserved OD shift in both >PD400 and >PD700 EE-mice was mediated by increases in open eye responses in V1 (adult OD plasticity). When SC-mice were transferred to EE after PD110, OD plasticity was restored until PD922. Moreover, visual abilities tested by both optomotry and the visual water task and interindividual variability were not different between PD700 SC- and EE-mice. Taken together, EE raising enabled a lifelong OD plasticity but did not affect basic visual performance.
Collapse
Affiliation(s)
- Franziska Greifzu
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie and Bernstein Fokus Neurotechnologie, Georg-August-Universität Göttingen, Göttingen, Germany.
| | - Evgenia Kalogeraki
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie and Bernstein Fokus Neurotechnologie, Georg-August-Universität Göttingen, Göttingen, Germany; Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, Göttingen, Germany
| | - Siegrid Löwel
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie and Bernstein Fokus Neurotechnologie, Georg-August-Universität Göttingen, Göttingen, Germany; Sensory Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| |
Collapse
|
20
|
Gazit N, Vertkin I, Shapira I, Helm M, Slomowitz E, Sheiba M, Mor Y, Rizzoli S, Slutsky I. IGF-1 Receptor Differentially Regulates Spontaneous and Evoked Transmission via Mitochondria at Hippocampal Synapses. Neuron 2016; 89:583-97. [PMID: 26804996 PMCID: PMC4742535 DOI: 10.1016/j.neuron.2015.12.034] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 11/19/2015] [Accepted: 12/13/2015] [Indexed: 12/22/2022]
Abstract
The insulin-like growth factor-1 receptor (IGF-1R) signaling is a key regulator of lifespan, growth, and development. While reduced IGF-1R signaling delays aging and Alzheimer's disease progression, whether and how it regulates information processing at central synapses remains elusive. Here, we show that presynaptic IGF-1Rs are basally active, regulating synaptic vesicle release and short-term plasticity in excitatory hippocampal neurons. Acute IGF-1R blockade or transient knockdown suppresses spike-evoked synaptic transmission and presynaptic cytosolic Ca(2+) transients, while promoting spontaneous transmission and resting Ca(2+) level. This dual effect on transmitter release is mediated by mitochondria that attenuate Ca(2+) buffering in the absence of spikes and decrease ATP production during spiking activity. We conclude that the mitochondria, activated by IGF-1R signaling, constitute a critical regulator of information processing in hippocampal neurons by maintaining evoked-to-spontaneous transmission ratio, while constraining synaptic facilitation at high frequencies. Excessive IGF-1R tone may contribute to hippocampal hyperactivity associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Neta Gazit
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Irena Vertkin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Martin Helm
- Department of Neuro- and Sensory Physiology, Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain, European Neuroscience Institute, University of Göttingen Medical Center, 37075 Göttingen, Germany; International Max Planck Research School Molecular Biology, 37077 Göttingen, Germany
| | - Edden Slomowitz
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Maayan Sheiba
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Yael Mor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Silvio Rizzoli
- Department of Neuro- and Sensory Physiology, Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain, European Neuroscience Institute, University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
21
|
Chumak T, Rüttiger L, Lee SC, Campanelli D, Zuccotti A, Singer W, Popelář J, Gutsche K, Geisler HS, Schraven SP, Jaumann M, Panford-Walsh R, Hu J, Schimmang T, Zimmermann U, Syka J, Knipper M. BDNF in Lower Brain Parts Modifies Auditory Fiber Activity to Gain Fidelity but Increases the Risk for Generation of Central Noise After Injury. Mol Neurobiol 2015; 53:5607-27. [PMID: 26476841 PMCID: PMC5012152 DOI: 10.1007/s12035-015-9474-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 10/05/2015] [Indexed: 11/24/2022]
Abstract
For all sensory organs, the establishment of spatial and temporal cortical resolution is assumed to be initiated by the first sensory experience and a BDNF-dependent increase in intracortical inhibition. To address the potential of cortical BDNF for sound processing, we used mice with a conditional deletion of BDNF in which Cre expression was under the control of the Pax2 or TrkC promoter. BDNF deletion profiles between these mice differ in the organ of Corti (BDNFPax2-KO) versus the auditory cortex and hippocampus (BDNFTrkC-KO). We demonstrate that BDNFPax2-KO but not BDNFTrkC-KO mice exhibit reduced sound-evoked suprathreshold ABR waves at the level of the auditory nerve (wave I) and inferior colliculus (IC) (wave IV), indicating that BDNF in lower brain regions but not in the auditory cortex improves sound sensitivity during hearing onset. Extracellular recording of IC neurons of BDNFPax2 mutant mice revealed that the reduced sensitivity of auditory fibers in these mice went hand in hand with elevated thresholds, reduced dynamic range, prolonged latency, and increased inhibitory strength in IC neurons. Reduced parvalbumin-positive contacts were found in the ascending auditory circuit, including the auditory cortex and hippocampus of BDNFPax2-KO, but not of BDNFTrkC-KO mice. Also, BDNFPax2-WT but not BDNFPax2-KO mice did lose basal inhibitory strength in IC neurons after acoustic trauma. These findings suggest that BDNF in the lower parts of the auditory system drives auditory fidelity along the entire ascending pathway up to the cortex by increasing inhibitory strength in behaviorally relevant frequency regions. Fidelity and inhibitory strength can be lost following auditory nerve injury leading to diminished sensory outcome and increased central noise.
Collapse
Affiliation(s)
- Tetyana Chumak
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Lukas Rüttiger
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Sze Chim Lee
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Dario Campanelli
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Annalisa Zuccotti
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany.,Department of Clinical Neurobiology, University Hospital and DKFZ Heidelberg, In Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Wibke Singer
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Jiří Popelář
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Katja Gutsche
- Instituto de Biologíay Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, E-47003, Valladolid, Spain
| | - Hyun-Soon Geisler
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Sebastian Philipp Schraven
- Department of Otolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Comprehensive Hearing Center, University of Würzburg, Josef-Schneider-Straße 11, 97080, Würzburg, Germany
| | - Mirko Jaumann
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | | | - Jing Hu
- Centre for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Straße 25, 72076, Tübingen, Germany
| | - Thomas Schimmang
- Instituto de Biologíay Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, E-47003, Valladolid, Spain
| | - Ulrike Zimmermann
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Josef Syka
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Marlies Knipper
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany.
| |
Collapse
|
22
|
Knipper M, Panford-Walsh R, Singer W, Rüttiger L, Zimmermann U. Specific synaptopathies diversify brain responses and hearing disorders: you lose the gain from early life. Cell Tissue Res 2015; 361:77-93. [PMID: 25843689 PMCID: PMC4487345 DOI: 10.1007/s00441-015-2168-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/05/2015] [Indexed: 01/08/2023]
Abstract
Before hearing onset, inner hair cell (IHC) maturation proceeds under the influence of spontaneous Ca(2+) action potentials (APs). The temporal signature of the IHC Ca(2+) AP is modified through an efferent cholinergic feedback from the medial olivocochlear bundle (MOC) and drives the IHC pre- and post-synapse phenotype towards low spontaneous (spike) rate (SR), high-threshold characteristics. With sensory experience, the IHC pre- and post-synapse phenotype matures towards the instruction of low-SR, high-threshold and of high-SR, low-threshold auditory fiber characteristics. Corticosteroid feedback together with local brain-derived nerve growth factor (BDNF) and catecholaminergic neurotransmitters (dopamine) might be essential for this developmental step. In this review, we address the question of whether the control of low-SR and high-SR fiber characteristics is linked to various degrees of vulnerability of auditory fibers in the mature system. In particular, we examine several IHC synaptopathies in the context of various hearing disorders and exemplified shortfalls before and after hearing onset.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | | | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | - Ulrike Zimmermann
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| |
Collapse
|
23
|
Li G, Xue Q, Luo Y, Hu X, Yu B. S6 inhibition contributes to isoflurane neurotoxicity in the developing brain. Toxicol Lett 2015; 233:102-13. [PMID: 25597859 DOI: 10.1016/j.toxlet.2014.11.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/18/2014] [Accepted: 11/21/2014] [Indexed: 02/07/2023]
Abstract
Postnatal isoflurane exposure leads to neurodegeneration and deficits of spatial learning and memory in the adulthood. However, the underlying mechanisms remain unclear. Ribosomal protein S6 is demonstrated to play a pivotal role in control of cell survival, protein synthesis and synaptogenesis for brain development. In this study, the possible role of S6 and its upstream signaling pathways in the developmental neurotoxicity of isoflurane was evaluated using models of primary cultured hippocampal neurons and postnatal day 7 rats. We found that isoflurane decreased IGF-1 level and suppressed activation of IGF-1 receptor, sequentially inhibiting S6 activity via IGF-1/MEK/ERK and IGF-1/PI3K/Akt signaling pathways. S6 inhibition enhanced isoflurane-induced decreased Bcl-xL and increased cleaved caspase-3 and Bad, also reduced PSD95 expression and aggravated deficits of spatial learning and memory. S6 activation could reverse the damages above. These results indicate that S6 inhibition, led by suppression of upstream IGF-1/MEK/ERK and IGF-1/PI3K/Akt signaling pathways, is involved in the neuroapoptosis, synaptogenesis impairment and spatial learning and memory decline caused by postnatal isoflurane exposure. S6 activation may exhibit protective potential against developmental neurotoxicity of isoflurane.
Collapse
Affiliation(s)
- Guohui Li
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Qingsheng Xue
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaodong Hu
- Department of Anatomy, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Buwei Yu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
24
|
O'Connor AM, Burton TJ, Leamey CA, Sawatari A. The use of the puzzle box as a means of assessing the efficacy of environmental enrichment. J Vis Exp 2014:52225. [PMID: 25590345 PMCID: PMC4354494 DOI: 10.3791/52225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Environmental enrichment can dramatically influence the development and function of neural circuits. Further, enrichment has been shown to successfully delay the onset of symptoms in models of Huntington's disease (1-4), suggesting environmental factors can evoke a neuroprotective effect against the progressive, cellular level damage observed in neurodegenerative disorders. The ways in which an animal can be environmentally enriched, however, can vary considerably. Further, there is no straightforward manner in which the effects of environmental enrichment can be assessed: most methods require either fairly complicated behavioral paradigms and/or postmortem anatomical/physiological analyses. This protocol describes the use of a simple and inexpensive behavioral assay, the Puzzle Box (5-7) as a robust means of determining the efficacy of increased social, sensory and motor stimulation on mice compared to cohorts raised in standard laboratory conditions. This simple problem solving task takes advantage of a rodent's innate desire to avoid open enclosures by seeking shelter. Cognitive ability is assessed by adding increasingly complex impediments to the shelter's entrance. The time a given subject takes to successfully remove the obstructions and enter the shelter serves as the primary metric for task performance. This method could provide a reliable means of rapidly assessing the efficacy of different enrichment protocols on cognitive function, thus paving the way for systematically determining the role specific environmental factors play in delaying the onset of neurodevelopmental and neurodegenerative disease.
Collapse
Affiliation(s)
| | - Thomas J Burton
- Discipline of Physiology, University of Sydney; Bosch Animal Behavioural Facility, University of Sydney
| | | | | |
Collapse
|
25
|
Barone I, Novelli E, Strettoi E. Long-term preservation of cone photoreceptors and visual acuity in rd10 mutant mice exposed to continuous environmental enrichment. Mol Vis 2014; 20:1545-56. [PMID: 25489227 PMCID: PMC4225138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/03/2014] [Indexed: 11/23/2022] Open
Abstract
PURPOSE In human patients and animal models of retinitis pigmentosa (RP), a gradual loss of rod photoreceptors and decline in scotopic vision are the primary manifestations of the disease. Secondary death of cones and gradual, regressive remodeling of the inner retina follow and progress at different speeds according to the underlying genetic defect. In any case, the final outcome is near-blindness without a conclusive cure yet. We recently reported that environmental enrichment (EE), an experimental manipulation based on exposure to enhanced motor, sensory, and social stimulation, when started at birth, exerts clear beneficial effects on a mouse model of RP, by slowing vision loss. The purpose of this study was to investigate in the same mouse the long-term effects of chronic exposure to an EE and assess the outcome of this manipulation on cone survival, inner retinal preservation, and visual behavior. METHODS Two groups of rd10 mutant mice were maintained in an EE or standard (ST) laboratory conditions up to 1 year of age. Then, retinal preservation was assessed with immunocytochemistry, confocal microscopy examination, cone counts, and electron microscopy of the photoreceptor layer, while visual acuity was tested behaviorally with a Prusky water maze. RESULTS rd10 mice are a model of autosomal recessive RP with a typical rod-cone, center to the periphery pattern of photoreceptor degeneration. They carry a mutation of the rod-specific phosphodiesterase gene and undergo rod death that peaks at around P24, while cone electroretinogram (ERG) is extinct by P60. We previously showed that early exposure to an EE efficiently delays photoreceptor degeneration in these mutants, extending the time window of cone viability and cone-mediated vision well beyond the phase of maximum rod death. Here we find that a maintained EE can delay the degeneration of cones even in the long term. Confocal and electron microscopy examination of the retinas of the rd10 EE and ST mice at 1 year of age showed major degeneration of the photoreceptor layer in both experimental groups, with small clusters of photoreceptors persisting in the peripheral retina. These vestigial cells were positive for L and M opsins and cone arrestin and represented the residual population of cones. In the retinas of the EE mice, cones were more numerous and less remodeled than in the ST counterparts, albeit virtually devoid of outer segments, as confirmed with electron microscopy (EM) observations. Cone counting in retinal whole mounts showed that rd10 EE mice at 1 year had almost three times as many surviving cones (34,000±4,000) as the ST control mice (12,700±1,800), t test p=0.003. Accordingly, the rd10 EE mice at 1 year of age were still capable of performing the visual water task in photopic conditions, showing a residual visual acuity of 0.138±0 cycles/degree. This ability was virtually absent in the rd10 ST age-matched mice (0.063±0.014), t test, p=0.029. No major differences were detected in the morphology of the neurons of the inner retina between the two experimental groups. CONCLUSIONS The approaches used to test the effects of an EE were consistent in showing significantly better preservation of cones and measurable visual acuity in 1-year-old rd10 EE mice. We therefore confirm and extend previous findings that showed an EE is an effective, minimally invasive tool for promoting long-lasting retinal protection in experimental models of RP.
Collapse
|
26
|
Spielman LJ, Little JP, Klegeris A. Inflammation and insulin/IGF-1 resistance as the possible link between obesity and neurodegeneration. J Neuroimmunol 2014; 273:8-21. [PMID: 24969117 DOI: 10.1016/j.jneuroim.2014.06.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 06/03/2014] [Accepted: 06/04/2014] [Indexed: 12/17/2022]
Abstract
Obesity is a growing epidemic that contributes to several brain disorders including Alzheimer's, Parkinson's, and Huntington's diseases. Obesity could promote these diseases through several different mechanisms. Here we review evidence supporting the involvement of two recently recognized factors linking obesity with neurodegeneration: the induction of pro-inflammatory cytokines and onset of insulin and insulin-like growth factor 1 (IGF-1) resistance. Excess peripheral pro-inflammatory mediators, some of which can cross the blood brain barrier, may trigger neuroinflammation, which subsequently exacerbates neurodegeneration. Insulin and IGF-1 resistance leads to weakening of neuroprotective signaling by these molecules and can contribute to onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lindsay J Spielman
- Department of Biology, University of British Columbia Okanagan Campus, 3333 University Way, Kelowna, BC, V1V 1V7 Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan Campus, 3333 University Way, Kelowna, BC, V1V 1V7 Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, 3333 University Way, Kelowna, BC, V1V 1V7 Canada.
| |
Collapse
|
27
|
Is there a relationship between brain-derived neurotrophic factor for driving neuronal auditory circuits with onset of auditory function and the changes following cochlear injury or during aging? Neuroscience 2014; 283:26-43. [PMID: 25064058 DOI: 10.1016/j.neuroscience.2014.07.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 01/06/2023]
Abstract
Brain-derived neurotrophic factor, BDNF, is one of the most important neurotrophic factors acting in the peripheral and central nervous system. In the auditory system its function was initially defined by using constitutive knockout mouse mutants and shown to be essential for survival of neurons and afferent innervation of hair cells in the peripheral auditory system. Further examination of BDNF null mutants also revealed a more complex requirement during re-innervation processes involving the efferent system of the cochlea. Using adult mouse mutants defective in BDNF signaling, it could be shown that a tonotopical gradient of BDNF expression within cochlear neurons is required for maintenance of a specific spatial innervation pattern of outer hair cells and inner hair cells. Additionally, BDNF is required for maintenance of voltage-gated potassium channels (KV) in cochlear neurons, which may form part of a maturation step within the ascending auditory pathway with onset of hearing and might be essential for cortical acuity of sound-processing and experience-dependent plasticity. A presumptive harmful role of BDNF during acoustic trauma and consequences of a loss of cochlear BDNF during aging are discussed in the context of a partial reversion of this maturation step. We compare the potentially beneficial and harmful roles of BDNF for the mature auditory system with those BDNF functions known in other sensory circuits, such as the vestibular, visual, olfactory, or somatosensory system.
Collapse
|
28
|
Sale A, Berardi N, Maffei L. Environment and Brain Plasticity: Towards an Endogenous Pharmacotherapy. Physiol Rev 2014; 94:189-234. [DOI: 10.1152/physrev.00036.2012] [Citation(s) in RCA: 265] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Brain plasticity refers to the remarkable property of cerebral neurons to change their structure and function in response to experience, a fundamental theoretical theme in the field of basic research and a major focus for neural rehabilitation following brain disease. While much of the early work on this topic was based on deprivation approaches relying on sensory experience reduction procedures, major advances have been recently obtained using the conceptually opposite paradigm of environmental enrichment, whereby an enhanced stimulation is provided at multiple cognitive, sensory, social, and motor levels. In this survey, we aim to review past and recent work concerning the influence exerted by the environment on brain plasticity processes, with special emphasis on the underlying cellular and molecular mechanisms and starting from experimental work on animal models to move to highly relevant work performed in humans. We will initiate introducing the concept of brain plasticity and describing classic paradigmatic examples to illustrate how changes at the level of neuronal properties can ultimately affect and direct key perceptual and behavioral outputs. Then, we describe the remarkable effects elicited by early stressful conditions, maternal care, and preweaning enrichment on central nervous system development, with a separate section focusing on neurodevelopmental disorders. A specific section is dedicated to the striking ability of environmental enrichment and physical exercise to empower adult brain plasticity. Finally, we analyze in the last section the ever-increasing available knowledge on the effects elicited by enriched living conditions on physiological and pathological aging brain processes.
Collapse
Affiliation(s)
- Alessandro Sale
- Institute of Neuroscience, National Research Council, Pisa, Italy; Department of Psychology, Florence University, Florence, Italy; and Scuola Normale Superiore, Pisa, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience, National Research Council, Pisa, Italy; Department of Psychology, Florence University, Florence, Italy; and Scuola Normale Superiore, Pisa, Italy
| | - Lamberto Maffei
- Institute of Neuroscience, National Research Council, Pisa, Italy; Department of Psychology, Florence University, Florence, Italy; and Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
29
|
Environmental enrichment decreases asphyxia-induced neurobehavioral developmental delay in neonatal rats. Int J Mol Sci 2013; 14:22258-73. [PMID: 24232451 PMCID: PMC3856064 DOI: 10.3390/ijms141122258] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/18/2013] [Accepted: 10/28/2013] [Indexed: 01/01/2023] Open
Abstract
Perinatal asphyxia during delivery produces long-term disability and represents a major problem in neonatal and pediatric care. Numerous neuroprotective approaches have been described to decrease the effects of perinatal asphyxia. Enriched environment is a popular strategy to counteract nervous system injuries. The aim of the present study was to investigate whether enriched environment is able to decrease the asphyxia-induced neurobehavioral developmental delay in neonatal rats. Asphyxia was induced in ready-to-deliver mothers by removing the pups by caesarian section after 15 min of asphyxia. Somatic and neurobehavioral development was tested daily and motor coordination weekly. Our results show that rats undergoing perinatal asphyxia had a marked developmental delay and worse performance in motor coordination tests. However, pups kept in enriched environment showed a decrease in the developmental delay observed in control asphyctic pups. Rats growing up in enriched environment did not show decrease in weight gain after the first week and the delay in reflex appearance was not as marked as in control rats. In addition, the development of motor coordination was not as strikingly delayed as in the control group. Short-term neurofunctional outcome are known to correlate with long-term deficits. Our results thus show that enriched environment could be a powerful strategy to decrease the deleterious developmental effects of perinatal asphyxia.
Collapse
|
30
|
Enriched early life experiences reduce adult anxiety-like behavior in rats: a role for insulin-like growth factor 1. J Neurosci 2013; 33:11715-23. [PMID: 23843538 DOI: 10.1523/jneurosci.3541-12.2013] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Early life experiences can affect brain development, contributing to shape interindividual differences in stress vulnerability and anxiety-like behavior. In rodents, high levels of maternal care have long-lasting positive effects on the behavior of the offspring and stress response; post-weaning rearing in an enriched environment (EE) or massage counteract the negative effects of maternal separation or prenatal stressors. We recently found that insulin-like growth factor 1 (IGF-1) is a key mediator of early EE or massage on brain development. Whether early enrichment of experience can induce long-lasting effects on anxiety-like behavior and whether IGF-1 is involved in these effects is not known. We assessed anxiety-like behavior by means of the elevated plus maze in control adult rats and in adult rats subjected to early EE or to massage. We found that both EE and massage reduced adult anxiety-like behavior. Early IGF-1 systemic injections in rat pups reared in standard condition mimic the effects of EE and massage, reducing anxiety-like behavior in the adult; blocking early IGF-1 action in massaged and EE animals prevents massage and EE effects. In EE and IGF-1-treated animals, we assessed the hippocampal expression of glucocorticoid receptors (GRs) at postnatal day 12 (P12) and P60, finding a significantly higher GR expression at P60 for both treatments. These results suggest that IGF-1 could be involved in mediating the long-lasting effects of early life experiences on vulnerability/resilience to stress in adults.
Collapse
|
31
|
Horvath G, Reglodi D, Vadasz G, Farkas J, Kiss P. Exposure to enriched environment decreases neurobehavioral deficits induced by neonatal glutamate toxicity. Int J Mol Sci 2013; 14:19054-66. [PMID: 24065102 PMCID: PMC3794820 DOI: 10.3390/ijms140919054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 08/08/2013] [Accepted: 08/14/2013] [Indexed: 12/30/2022] Open
Abstract
Environmental enrichment is a popular strategy to enhance motor and cognitive performance and to counteract the effects of various harmful stimuli. The protective effects of enriched environment have been shown in traumatic, ischemic and toxic nervous system lesions. Monosodium glutamate (MSG) is a commonly used taste enhancer causing excitotoxic effects when given in newborn animals. We have previously demonstrated that MSG leads to a delay in neurobehavioral development, as shown by the delayed appearance of neurological reflexes and maturation of motor coordination. In the present study we aimed at investigating whether environmental enrichment is able to decrease the neurobehavioral delay caused by neonatal MSG treatment. Newborn pups were treated with MSG subcutaneously on postnatal days 1, 5 and 9. For environmental enrichment, we placed rats in larger cages, supplemented with different toys that were altered daily. Normal control and enriched control rats received saline treatment only. Physical parameters such as weight, day of eye opening, incisor eruption and ear unfolding were recorded. Animals were observed for appearance of reflexes such as negative geotaxis, righting reflexes, fore- and hindlimb grasp, fore- and hindlimb placing, sensory reflexes and gait. In cases of negative geotaxis, surface righting and gait, the time to perform the reflex was also recorded daily. For examining motor coordination, we performed grid walking, footfault, rope suspension, rota-rod, inclined board and walk initiation tests. We found that enriched environment alone did not lead to marked alterations in the course of development. On the other hand, MSG treatment caused a slight delay in reflex development and a pronounced delay in weight gain and motor coordination maturation. This delay in most signs and tests could be reversed by enriched environment: MSG-treated pups kept under enriched conditions showed no weight retardation, no reflex delay in some signs and performed better in most coordination tests. These results show that environmental enrichment is able to decrease the neurobehavioral delay caused by neonatal excitotoxicity.
Collapse
Affiliation(s)
- Gabor Horvath
- Department of Anatomy, PTE-MTA Lendulet PACAP Research Team, University of Pecs, Pécs 7624, Hungary.
| | | | | | | | | |
Collapse
|
32
|
Gender-dependent effects of enriched environment and social isolation in ischemic retinal lesion in adult rats. Int J Mol Sci 2013; 14:16111-23. [PMID: 23921682 PMCID: PMC3759902 DOI: 10.3390/ijms140816111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/10/2013] [Accepted: 07/17/2013] [Indexed: 12/17/2022] Open
Abstract
Exposure to an enriched environment has been shown to have many positive effects on brain structure and function. Numerous studies have proven that enriched environment can reduce the lesion induced by toxic and traumatic injuries. Impoverished environment, on the other hand, can have deleterious effects on the outcome of neuronal injuries. We have previously shown that enriched conditions have protective effects in retinal injury in newborn rats. It is well-known that the efficacy of neuroprotective strategies can depend on age and gender. The aim of the present study, therefore, was to examine the effects of environmental enrichment and social isolation in retinal ischemia. We used bilateral common carotid artery occlusion to induce retinal hypoperfusion in adult Wistar rats of both genders. Groups were housed in standard, enriched or impoverished conditions. Impoverished environment was induced by social isolation. Retinas were processed for histological analysis after two weeks of survival. In the present study, we show that (1) enriched environment has protective effects in adult ischemic retinal lesion, while (2) impoverished environment further increases the degree of ischemic injury, and (3) that these environmental effects are gender-dependent: females are less responsive to the positive effects of environmental enrichment and more vulnerable to retinal ischemia in social isolation. In summary, our present study shows that the effects of both positive and negative environmental stimuli are gender-dependent in ischemic retinal lesions.
Collapse
|
33
|
System consolidation of spatial memories in mice: effects of enriched environment. Neural Plast 2013; 2013:956312. [PMID: 23936679 PMCID: PMC3723323 DOI: 10.1155/2013/956312] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 06/15/2013] [Indexed: 01/28/2023] Open
Abstract
Environmental enrichment (EE) is known to enhance learning and memory. Declarative memories are thought to undergo a first rapid and local consolidation process, followed by a prolonged process of system consolidation, which consist in a time-dependent gradual reorganization of brain regions supporting remote memory storage and crucial for the formation of enduring memories. At present, it is not known whether EE can affect the process of declarative memory system consolidation. We characterized the time course of hippocampal and cortical activation following recall of progressively more remote spatial memories. Wild-type mice either exposed to EE for 40 days or left in standard environment were subjected to spatial learning in the Morris water maze and to the probe test 1, 10, 20, 30, and 50 days after learning. Following the probe test, regional expression of the inducible immediate early gene c-Fos was mapped by immunohistochemistry, as an indicator of neuronal activity. We found that activation of the medial prefrontal cortex (mPFC), suggested to have a privileged role in processing remote spatial memories, was evident at shorter time intervals after learning in EE mice; in addition, EE induced the progressive activation of a distributed cortical network not activated in non-EE mice. This suggests that EE not only accelerates the process of mPFC recruitment but also recruits additional cortical areas into the network supporting remote spatial memories.
Collapse
|
34
|
Cyclic AMP-dependent regulation of tyrosine hydroxylase mRNA and immunofluorescence levels in rat retinal precursor cells. Cell Tissue Res 2013; 352:207-16. [PMID: 23355011 DOI: 10.1007/s00441-013-1555-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 01/03/2013] [Indexed: 10/27/2022]
Abstract
Stimulation of tyrosine hydroxylase (TH) gene transcription by cyclic AMP (cAMP) has been clearly established in adrenal medula cells and neural-crest-derived cell lines but information on this mechanism is still lacking in dopaminergic neurons. Because they are easily amenable to in vitro experiments, dopaminergic amacrine cells of the retina might constitute a valuable model system to study this mechanism. We have used real-time reverse transcription with the polymerase chain reaction to quantify TH mRNA levels in the rat retina during post-natal development and in retinal precursor cells obtained from neonatal rats and cultured for 3 days in serum-free medium. Whereas the TH mRNA concentration remains consistantly low in control cultures, treatment with cAMP-increasing agents (forskolin, membrane depolarization, phosphodiesterase inhibitors) is sufficient to raise it to the level observed in adult retina (15-fold increase). Treatment of the cultured cells can be delayed by up to 2 days with identical results at the TH mRNA level, thus ruling out a survival-promoting effect of cAMP. TH immunofluorescence has confirmed cAMP-dependent regulation of TH expression at the protein level and indicates that the frequency of TH-positive cells in the cultures is similar to that observed in the adult retina. Selective phosphodiesterase inhibitors suggest that PDE4 is the major subtype involved in the dopaminergic amacrine cell response. Our data clearly establish the cAMP-dependent regulation of TH mRNA and immunofluorescence levels in retinal precursor cells. The possible role of this regulation mechanism in the developmental activation of TH gene expression is discussed.
Collapse
|
35
|
Abstract
Brain development in neurodevelopmental disorders has been considered to comprise a sequence of critical periods, and abnormalities occurring during early development have been considered irreversible in adulthood. However, findings in mouse models of neurodevelopmental disorders, including fragile X, Rett syndrome, Down syndrome, and neurofibromatosis type I suggest that it is possible to reverse certain molecular, electrophysiological, and behavioral deficits associated with these disorders in adults by genetic or pharmacological manipulations. Furthermore, recent studies have suggested that critical period-like plasticity can be reactivated in the adult brain by environmental manipulations or by pharmacotherapy. These studies open up a tantalizing possibility that targeted pharmacological treatments in combination with regimes of training or rehabilitation might alleviate or reverse the symptoms of neurodevelopmental disorders even after the end of critical developmental periods. Even though translation from animal experimentation to clinical practice is challenging, these results suggest a rational basis for treatment of neurodevelopmental disorders in adulthood.
Collapse
|
36
|
Barone I, Novelli E, Piano I, Gargini C, Strettoi E. Environmental enrichment extends photoreceptor survival and visual function in a mouse model of retinitis pigmentosa. PLoS One 2012; 7:e50726. [PMID: 23209820 PMCID: PMC3508993 DOI: 10.1371/journal.pone.0050726] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 10/24/2012] [Indexed: 01/03/2023] Open
Abstract
Slow, progressive rod degeneration followed by cone death leading to blindness is the pathological signature of all forms of human retinitis pigmentosa (RP). Therapeutic schemes based on intraocular delivery of neuroprotective agents prolong the lifetime of photoreceptors and have reached the stage of clinical trial. The success of these approaches depends upon optimization of chronic supply and appropriate combination of factors. Environmental enrichment (EE), a novel neuroprotective strategy based on enhanced motor, sensory and social stimulation, has already been shown to exert beneficial effects in animal models of various disorders of the CNS, including Alzheimer and Huntington disease. Here we report the results of prolonged exposure of rd10 mice, a mutant strain undergoing progressive photoreceptor degeneration mimicking human RP, to such an enriched environment from birth. By means of microscopy of retinal tissue, electrophysiological recordings, visual behaviour assessment and molecular analysis, we show that EE considerably preserves retinal morphology and physiology as well as visual perception over time in rd10 mutant mice. We find that protective effects of EE are accompanied by increased expression of retinal mRNAs for CNTF and mTOR, both factors known as instrumental to photoreceptor survival. Compared to other rescue approaches used in similar animal models, EE is highly effective, minimally invasive and results into a long-lasting retinal protection. These results open novel perspectives of research pointing to environmental strategies as useful tools to extend photoreceptor survival.
Collapse
Affiliation(s)
- Ilaria Barone
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, Italy
| | | | | | | | | |
Collapse
|
37
|
Wen Z, Zeng W, Dai J, Zhou X, Yang C, Duan F, Liu Y, Yang H, Yuan L. Paravertebral fascial massage promotes brain development of neonatal rats via the insulin-like growth factor 1 pathway. Neural Regen Res 2012; 7:1185-91. [PMID: 25722713 PMCID: PMC4340037 DOI: 10.3969/j.issn.1673-5374.2012.15.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/20/2012] [Indexed: 01/31/2023] Open
Abstract
Massage in traditional Chinese medicine can promote body and brain development of premature and normal newborn infants. In the present study, neonatal rats (1 day old) underwent paravertebral fascial massage (15 consecutive days), followed by subcutaneous injection of insulin-like growth factor 1 receptor antagonist, JB1 (9 consecutive days). Paravertebral fascial massage significantly increased insulin-like growth factor 1 expression and cell proliferation in the subventricular zone of the lateral ventricle and dentate gyrus of the hippocampus. However, JB1 inhibited this increase. Results suggest that paravertebral fascial massage can promote brain development of neonatal rats via the insulin-like growth factor 1 pathway.
Collapse
Affiliation(s)
- Zhongqiu Wen
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Wenqin Zeng
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Jingxing Dai
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xin Zhou
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Chun Yang
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Fuhua Duan
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Yufeng Liu
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Huiying Yang
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Lin Yuan
- Key Laboratory of Tissue Construction and Detection of Guangdong Province, Department of Anatomy, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
38
|
West EL, Pearson RA, Duran Y, Gonzalez-Cordero A, MacLaren RE, Smith AJ, Sowden JC, Ali RR. Manipulation of the recipient retinal environment by ectopic expression of neurotrophic growth factors can improve transplanted photoreceptor integration and survival. Cell Transplant 2012; 21:871-87. [PMID: 22325046 DOI: 10.3727/096368911x623871] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Degeneration of the neural retina is the leading cause of untreatable blindness in the developed world. Stem cell replacement therapy offers a novel strategy for retinal repair. Postmitotic photoreceptor precursors derived from the early postnatal (P) retina are able to migrate and integrate into the adult mouse retina following transplantation into the subretinal space, but it is likely that a large number of these cells would be required to restore vision. The adult recipient retina presents a very different environment to that from which photoreceptor precursor donor cells isolated from the developing postnatal retina are derived. Here we considered the possibility that modulation of the recipient environment by ectopic expression of developmentally regulated growth factors, normally present during photoreceptor development, might enhance the migration and integration of transplanted cells into the adult neural retina. Adeno-associated viral (AAV) vectors were used to introduce three growth factors previously reported to play a role in photoreceptor development, IGF1, FGF2, and CNTF, into the adult retina, prior to transplantation of P4 cells derived from the Nrl.GFP(+ve) neural retina. At 3 weeks posttransplantation the number of integrated, differentiated photoreceptor cells present in AAV-mediated neurotrophic factor-treated eyes was assessed and compared to control treated contralateral eyes. We show, firstly, that it is possible to manipulate the recipient retinal microenvironment via rAAV-mediated gene transfer with respect to these developmentally relevant growth factors. Moreover, when combined with cell transplantation, AAV-mediated expression of IGF1 led to significantly increased levels of cell integration, while overexpression of FGF2 had no significant effect on integrated cell number. Conversely, expression of CNTF led to a significant decrease in cell integration and an exacerbated glial response that led to glial scarring. Together, these findings demonstrate the importance of the extrinsic environment of the recipient retina for photoreceptor cell transplantation and show for the first time that it is possible to manipulate this environment using viral vectors to influence photoreceptor transplantation efficiency.
Collapse
Affiliation(s)
- E L West
- Department of Genetics, University College London Institute of Ophthalmology, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Overexpression of neurotrophin-3 stimulates a second wave of dopaminergic amacrine cell genesis after birth in the mouse retina. J Neurosci 2011; 31:12663-73. [PMID: 21880927 DOI: 10.1523/jneurosci.1100-11.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dopaminergic amacrine (DA) cells play multiple and important roles in retinal function. Neurotrophins are known to modulate the number and morphology of DA cells, but the underlying regulatory mechanisms are unclear. Here, we investigate how neurotrophin-3 (NT-3) regulates DA cell density in the mouse retina. We demonstrate that overexpression of NT-3 upregulates DA cell number and leads to a consequent increase in the density of DA cell dendrites. To examine the mechanisms of DA cell density increase, we further investigate the effect of NT-3 overexpression on retinal apoptosis and mitosis during development. We find that NT-3 does not affect the well known wave of retinal cell apoptosis that normally occurs during the first 2 weeks after birth. Instead, overexpression of NT-3 promotes additional mitosis of DA cells at postnatal day 4, but does not affect cell mitosis before birth, the peak period of amacrine cell genesis in wild-type retinas. We next show that retinal explants cultured from birth to day 7 without extra NT-3 produced by lens exhibit similar number of DA cells as in wild type, further supporting the notion that postnatal overexpression of lens-derived NT-3 affects DA cell number. Moreover, the additional mitosis after birth in NT-3-overexpressing mice does not occur in calretinin-positive amacrine cells or PKC-positive rod ON bipolar cells. Thus, the NT-3-triggered wave of cell mitosis after birth is specific for the retinal DA cells.
Collapse
|
40
|
Takuma K, Ago Y, Matsuda T. Preventive effects of an enriched environment on rodent psychiatric disorder models. J Pharmacol Sci 2011; 117:71-6. [PMID: 21881295 DOI: 10.1254/jphs.11r07cp] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Interplay between genetic and environmental factors plays a key role in psychiatric disorders, as well as other brain diseases, cancer, and metabolic syndrome. In accordance with epidemiological findings, animal studies have pointed out the importance of a variety of environmental factors, such as viral infection during pregnancy or infancy, early parental loss or separation, and physical or sexual abuse in early life, in the etiology of psychiatric disorders. Conversely, positive effects of environmental factors against the pathogenesis of psychiatric disorders are also demonstrated, in which most of the animals are exposed to an "enriched environment". This review summarizes recent progress of research in this field focusing on the preventive effects of an "enriched environment" against the expression of behavioral abnormalities in rodent models of psychiatric disorders.
Collapse
Affiliation(s)
- Kazuhiro Takuma
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | | | | |
Collapse
|
41
|
Landi S, Putignano E, Boggio EM, Giustetto M, Pizzorusso T, Ratto GM. The short-time structural plasticity of dendritic spines is altered in a model of Rett syndrome. Sci Rep 2011; 1:45. [PMID: 22355564 PMCID: PMC3216532 DOI: 10.1038/srep00045] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 07/04/2011] [Indexed: 11/09/2022] Open
Abstract
The maturation of excitatory transmission comes about through a developmental period in which dendritic spines are highly motile and their number, form and size are rapidly changing. Surprisingly, although these processes are crucial for the formation of cortical circuitry, little is known about possible alterations of these processes in brain disease. By means of acute in vivo 2-photon imaging we show that the dynamic properties of dendritic spines of layer V cortical neurons are deeply affected in a mouse model of Rett syndrome (RTT) at a time around P25 when the neuronal phenotype of the disease is still mild. Then, we show that 24h after a subcutaneous injection of IGF-1 spine dynamics is restored. Our study demonstrates that spine dynamics in RTT mice is severely impaired early during development and suggest that treatments for RTT should be started very early in order to reestablish a normal period of spine plasticity.
Collapse
Affiliation(s)
- Silvia Landi
- NEST, Scuola Normale Superiore, Pisa, Italy; NEST Institute Nanoscience CNR, Pisa, Italy
| | | | | | | | | | | |
Collapse
|
42
|
Mainardi M, Landi S, Gianfranceschi L, Baldini S, De Pasquale R, Berardi N, Maffei L, Caleo M. Environmental enrichment potentiates thalamocortical transmission and plasticity in the adult rat visual cortex. J Neurosci Res 2011; 88:3048-59. [PMID: 20722076 DOI: 10.1002/jnr.22461] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
It has been demonstrated that the complex sensorimotor and social stimulation achieved by rearing animals in an enriched environment (EE) can reinstate juvenile-like plasticity in the adult cortex. However, it is not known whether EE can affect thalamocortical transmission. Here, we recorded in vivo field potentials from the visual cortex evoked by electrical stimulation of the dorsal lateral geniculate nucleus (dLGN) in anesthetized rats. We found that a period of EE during adulthood shifted the input-output curves and increased paired-pulse depression, suggesting an enhanced synaptic strength at thalamocortical terminals. Accordingly, EE animals showed an increased expression of the vesicular glutamate transporter 2 (vGluT-2) in geniculocortical afferents to layer IV. Rats reared in EE also showed an enhancement of thalamocortical long-term potentiation (LTP) triggered by theta-burst stimulation (TBS) of the dLGN. To monitor the functional consequences of increased LTP in EE rats, we recorded visual evoked potentials (VEPs) before and after application of TBS to the geniculocortical pathway. We found that responses to visual stimulation were enhanced across a range of contrasts in EE animals. This was accompanied by an up-regulation of the intracortical excitatory synaptic marker vGluT-1 and a decrease in the expression of the vesicular GABA transporter (vGAT), indicating a shift in the excitation/inhibition ratio. Thus, in the adult rat, EE enhances synaptic strength and plasticity of the thalamocortical pathway associated with specific changes in glutamatergic and GABAergic neurotransmission. These data provide novel insights into the mechanisms by which EE shapes the adult brain.
Collapse
Affiliation(s)
- Marco Mainardi
- Laboratory of Neurobiology, Scuola Normale Superiore, Pisa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
IGF-1 (insulin-like growth factor-1) plays a unique role in the cell protection of multiple systems, where its fine-tuned signal transduction helps to preserve tissues from hypoxia, ischaemia and oxidative stress, thus mediating functional homoeostatic adjustments. In contrast, its deprivation results in apoptosis and dysfunction. Many prospective epidemiological surveys have associated low IGF-1 levels with late mortality, MI (myocardial infarction), HF (heart failure) and diabetes. Interventional studies suggest that IGF-1 has anti-atherogenic actions, owing to its multifaceted impact on cardiovascular risk factors and diseases. The metabolic ability of IGF-1 in coupling vasodilation with improved function plays a key role in these actions. The endothelial-protective, anti-platelet and anti-thrombotic activities of IGF-1 exert critical effects in preventing both vascular damage and mechanisms that lead to unstable coronary plaques and syndromes. The pro-survival and anti-inflammatory short-term properties of IGF-1 appear to reduce infarct size and improve LV (left ventricular) remodelling after MI. An immune-modulatory ability, which is able to suppress 'friendly fire' and autoreactivity, is a proposed important additional mechanism explaining the anti-thrombotic and anti-remodelling activities of IGF-1. The concern of cancer risk raised by long-term therapy with IGF-1, however, deserves further study. In the present review, we discuss the large body of published evidence and review data on rhIGF-1 (recombinant human IGF-1) administration in cardiovascular disease and diabetes, with a focus on dosage and safety issues. Perhaps the time has come for the regenerative properties of IGF-1 to be assessed as a new pharmacological tool in cardiovascular medicine.
Collapse
|
44
|
Kiss P, Atlasz T, Szabadfi K, Horvath G, Griecs M, Farkas J, Matkovits A, Toth G, Lubics A, Tamas A, Gabriel R, Reglodi D. Comparison between PACAP- and enriched environment-induced retinal protection in MSG-treated newborn rats. Neurosci Lett 2010; 487:400-5. [PMID: 21050880 DOI: 10.1016/j.neulet.2010.10.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 10/25/2010] [Accepted: 10/27/2010] [Indexed: 10/18/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) and its receptors occur throughout the nervous system, including the retina. PACAP exerts diverse actions in the eye: it influences ocular blood flow, contraction of the ciliary muscle, and has retinoprotective effects. This effect has been proven in different models of retinal degeneration. We have previously shown that PACAP protects against monosodium-glutamate (MSG)-induced damage in neonatal rats. The beneficial effects of enriched environment, another neuroprotective strategy, have long been known. Environmental enrichment has been shown to decrease different neuronal injuries. It also influences the development of the visual system. We have recently demonstrated that significant neuroprotection can be achieved in MSG-induced retinal degeneration in animals kept in an enriched environment. Combination of neuroprotective strategies often results in increased protection. Therefore, the aim of the present study was to compare the two neuroprotective strategies alone and in combination therapy. We found that both PACAP and environmental enrichment led to a similar degree of retinal protection, but the two treatments together did not lead to increased protection: their effects were not additive.
Collapse
Affiliation(s)
- Peter Kiss
- Department of Anatomy, University of Pecs, 7624 Pecs, Szigeti u 12, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cantrell DR, Cang J, Troy JB, Liu X. Non-centered spike-triggered covariance analysis reveals neurotrophin-3 as a developmental regulator of receptive field properties of ON-OFF retinal ganglion cells. PLoS Comput Biol 2010; 6:e1000967. [PMID: 20975932 PMCID: PMC2958799 DOI: 10.1371/journal.pcbi.1000967] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 09/21/2010] [Indexed: 01/11/2023] Open
Abstract
The functional separation of ON and OFF pathways, one of the fundamental features of the visual system, starts in the retina. During postnatal development, some retinal ganglion cells (RGCs) whose dendrites arborize in both ON and OFF sublaminae of the inner plexiform layer transform into RGCs with dendrites that monostratify in either the ON or OFF sublamina, acquiring final dendritic morphology in a subtype-dependent manner. Little is known about how the receptive field (RF) properties of ON, OFF, and ON-OFF RGCs mature during this time because of the lack of a reliable and efficient method to classify RGCs into these subtypes. To address this deficiency, we developed an innovative variant of Spike Triggered Covariance (STC) analysis, which we term Spike Triggered Covariance – Non-Centered (STC-NC) analysis. Using a multi-electrode array (MEA), we recorded the responses of a large population of mouse RGCs to a Gaussian white noise stimulus. As expected, the Spike-Triggered Average (STA) fails to identify responses driven by symmetric static nonlinearities such as those that underlie ON-OFF center RGC behavior. The STC-NC technique, in contrast, provides an efficient means to identify ON-OFF responses and quantify their RF center sizes accurately. Using this new tool, we find that RGCs gradually develop sensitivity to focal stimulation after eye opening, that the percentage of ON-OFF center cells decreases with age, and that RF centers of ON and ON-OFF cells become smaller. Importantly, we demonstrate for the first time that neurotrophin-3 (NT-3) regulates the development of physiological properties of ON-OFF center RGCs. Overexpression of NT-3 leads to the precocious maturation of RGC responsiveness and accelerates the developmental decrease of RF center size in ON-OFF cells. In summary, our study introduces STC-NC analysis which successfully identifies subtype RGCs and demonstrates how RF development relates to a neurotrophic driver in the retina. The developmental separation of ON and OFF pathways is one of the fundamental features of the visual system. In the mouse retina, some bi-stratified ON-OFF RGCs are refined into mono-stratified ON or OFF RGCs during the first postnatal month. However, the process by which the RGCs' physiological receptive field properties mature remains incompletely characterized, mainly due to the lack of a reliable and efficient method to classify RGCs into different subtypes. Here we have developed an innovative analysis, Spike Triggered Covariance – Non-Centered (STC-NC), and demonstrated that this technique can accurately characterize the receptive field properties of ON, OFF and ON-OFF center cells. We show that, in wildtype mouse, RGCs gradually develop sensitivity to focal stimulation after eye opening, and the development of ON-OFF receptive field center properties correlates well with their dendritic laminar refinement. Furthermore, overexpression of NT-3 accelerates the developmental decrease of receptive field center size in ON-OFF cells. Our study is the first to establish the STC-NC analysis which can successfully identify ON-OFF subtype RGCs and to demonstrate how receptive field development relates to a neurotrophic driver in the retina.
Collapse
Affiliation(s)
- Donald R. Cantrell
- Interdepartmental Neuroscience Program, Northwestern University, Evanston, Illinois, United States of America
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Jianhua Cang
- Interdepartmental Neuroscience Program, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - John B. Troy
- Interdepartmental Neuroscience Program, Northwestern University, Evanston, Illinois, United States of America
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
- * E-mail: (JBT); (XL)
| | - Xiaorong Liu
- Interdepartmental Neuroscience Program, Northwestern University, Evanston, Illinois, United States of America
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
- * E-mail: (JBT); (XL)
| |
Collapse
|
46
|
Abstract
Insulin-like growth factor I (IGF-I) belongs to an ancient family of hormones already present in early invertebrates. The insulin family is well characterized in mammals, although new members have been described recently. Since its characterization over 50 years ago, IGF-I has been considered a peptide mostly involved in the control of body growth and tissue remodeling. Currently, its most prominent recognized role is as a quasi-universal cytoprotectant. This role connects IGF-I with regulation of lifespan and with cancer, two areas of very active research in relation to this peptide. In the brain, IGF-I was formerly considered a neurotrophic factor involved in brain growth, as many other neurotrophic factors. Other aspects of the neurobiology of IGF-I are gradually emerging and suggest that this growth factor has a prominent role in brain function as a whole. During development IGF-I is abundantly expressed in many areas, whereas once the brain is formed its expression is restricted to a few regions and in very low quantities. However, the adult brain appears to have an external input from serum IGF-I, where this anabolic peptide is abundant. Thus, serum IGF-I has been proven to be an important modulator of brain activity, including higher functions such as cognition. Many of these functions can be ascribed to its tissue-remodeling activity as IGF-I modulates adult neurogenesis and angiogenesis. Other activities are cytoprotective; indeed, IGF-I can be considered a key neuroprotective peptide. Still others pertain to the functional characteristics of brain cells, such as cell excitability. Through modulation of membrane channels and neurotransmission, IGF-I impinges directly on neuronal plasticity, the cellular substrate of cognition. However, to fully understand the role of IGF-I in the brain, we have to sum the actions of locally produced IGF-I to those of serum IGF-I, and this is still pending. Thus, an integrated view of the role played by IGF-I in the brain is not yet possible. An operational approach to overcome this limitation would be to consider IGF-I as a signal coupling environmental influences on body metabolism with brain function. Or in a more colloquial way, we may say that IGF-I links body "fitness" with brain fitness, providing a mechanism to the roman saying "mens sana in corpore sano."
Collapse
|
47
|
Baroncelli L, Braschi C, Spolidoro M, Begenisic T, Sale A, Maffei L. Nurturing brain plasticity: impact of environmental enrichment. Cell Death Differ 2009; 17:1092-103. [PMID: 20019745 DOI: 10.1038/cdd.2009.193] [Citation(s) in RCA: 188] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Environmental enrichment (EE) is known to profoundly affect the central nervous system (CNS) at the functional, anatomical and molecular level, both during the critical period and during adulthood. Recent studies focusing on the visual system have shown that these effects are associated with the recruitment of previously unsuspected neural plasticity processes. At early stages of brain development, EE triggers a marked acceleration in the maturation of the visual system, with maternal behaviour acting as a fundamental mediator of the enriched experience in both the foetus and the newborn. In adult brain, EE enhances plasticity in the cerebral cortex, allowing the recovery of visual functions in amblyopic animals. The molecular substrate of the effects of EE on brain plasticity is multi-factorial, with reduced intracerebral inhibition, enhanced neurotrophin expression and epigenetic changes at the level of chromatin structure. These findings shed new light on the potential of EE as a non-invasive strategy to ameliorate deficits in the development of the CNS and to treat neurological disorders.
Collapse
Affiliation(s)
- L Baroncelli
- Laboratory of Neurobiology, Scuola Normale Superiore, Pisa I-56100, Italy.
| | | | | | | | | | | |
Collapse
|