1
|
Krishnamurthy R, Schultz DH, Wang Y, Natarajan SK, Barlow SM, Dietsch AM. Multimodal Adaptations to Expiratory Musculature-Targeted Resistance Training: A Preliminary Study in Healthy Young Adults. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2025; 68:987-1005. [PMID: 39908358 DOI: 10.1044/2024_jslhr-24-00294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
PURPOSE Exercise-induced adaptations, including neuroplasticity, are well studied for physical exercise that targets skeletal muscles. However, little is known about the neuroplastic potential of targeted speech and swallowing exercises. The current study aimed to gather preliminary data on molecular and functional changes associated with the neuroplastic effects of 4-week expiratory musculature-targeted resistance training in healthy young adults. METHOD Five healthy young adult men aged between 19 and 35 years, M (SD) = 28.8 (2.68) years, underwent 4 weeks of expiratory muscle strength training (EMST). We measured changes in maximum expiratory pressure (MEP), serum brain-derived neurotrophic factor (BDNF), and insulin-like growth factor 1 (IGF-1) levels at baseline and posttraining conditions. Furthermore, functional and structural magnetic resonance images were obtained to investigate the neuroplastic effects of EMST. We analyzed the effects of training using a linear mixed model for each outcome, with fixed effects for baseline and posttraining. RESULTS MEP and serum BDNF levels significantly increased posttraining. However, this effect was not observed for IGF-1. A significant increase in functional activation in eight regions was also observed posttraining. However, we did not observe significant changes in the white matter microstructure. CONCLUSIONS Preliminary data from our study suggest targeted resistance training of expiratory muscles results in molecular and neuroplastic adaptations similar to exercise that targets skeletal muscles. Additionally, these results suggest that EMST could be a potential intervention to modulate (or prime) neurotrophic signaling pathways linked to functional strength gains and neuroplasticity.
Collapse
Affiliation(s)
- Rahul Krishnamurthy
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln
| | - Douglas H Schultz
- Center for Brain, Biology, and Behavior, University of Nebraska-Lincoln
- Department of Psychology, University of Nebraska-Lincoln
| | - Yingying Wang
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln
- Center for Brain, Biology, and Behavior, University of Nebraska-Lincoln
| | | | - Steven M Barlow
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln
- Center for Brain, Biology, and Behavior, University of Nebraska-Lincoln
- Department of Biological Systems Engineering, University of Nebraska-Lincoln
| | - Angela M Dietsch
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln
- Center for Brain, Biology, and Behavior, University of Nebraska-Lincoln
| |
Collapse
|
2
|
Kostka M, Morys J, Małecki A, Nowacka-Chmielewska M. Muscle-brain crosstalk mediated by exercise-induced myokines - insights from experimental studies. Front Physiol 2024; 15:1488375. [PMID: 39687518 PMCID: PMC11647023 DOI: 10.3389/fphys.2024.1488375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Over the past couple of decades, it has become apparent that skeletal muscles might be engaged in endocrine signaling, mostly as a result of exercise or physical activity in general. The importance of this phenomenon is currently studied in terms of the impact that exercise- or physical activity -induced signaling factors have, in the interaction of the "muscle-brain crosstalk." So far, skeletal muscle-derived myokines were demonstrated to intercede in the connection between muscles and a plethora of various organs such as adipose tissue, liver, or pancreas. However, the exact mechanism of muscle-brain communication is yet to be determined. It is speculated that, in particular, brain-derived neurotrophic factor (BDNF), irisin, cathepsin B (CTSB), interleukin 6 (IL-6), and insulin-like growth factor-1 (IGF-1) partake in this crosstalk by promoting neuronal proliferation and synaptic plasticity, also resulting in improved cognition and ameliorated behavioral alterations. Researchers suggest that myokines might act directly on the brain parenchyma via crossing the blood-brain barrier (BBB). The following article reviews the information available regarding rodent studies on main myokines determined to cross the BBB, specifically addressing the association between exercise-induced myokine release and central nervous system (CNS) impairments. Although the hypothesis of skeletal muscles being critical sources of myokines seems promising, it should not be forgotten that the origin of these factors might vary, depending on the cell types engaged in their synthesis. Limited amount of research providing information on alterations in myokines expression in various organs at the same time, results in taking them only as circumstantial evidence on the way to determine the actual involvement of skeletal muscles in the overall state of homeostasis. The following article reviews the information available regarding rodent studies on main myokines determined to cross the BBB, specifically addressing the association between exercise-induced myokine release and CNS impairments.
Collapse
Affiliation(s)
| | | | | | - Marta Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
| |
Collapse
|
3
|
Walser M, Karlsson L, Motalleb R, Isgaard J, Kuhn HG, Svensson J, Åberg ND. Running in mice increases the expression of brain hemoglobin-related genes interacting with the GH/IGF-1 system. Sci Rep 2024; 14:25464. [PMID: 39462081 PMCID: PMC11513053 DOI: 10.1038/s41598-024-77489-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
The beneficial effects of exercise are partly mediated via local or systemic functions of the insulin-like growth factor-1 (IGF-1) system. As IGF-1 increases local brain hemoglobin beta (Hbb) transcripts, we hypothesized that exercise could have similar effects. Mice were single-housed with free access to running wheels for seven days. After sacrifice and saline perfusion, the expression of 13 genes was quantified using real-time quantitative polymerase chain reaction (RT-qPCR) in three brain regions: the prefrontal cortex, motor cortex, and hippocampus. In addition, plasma insulin, glucose, homeostatic model assessment of IR (HOMA-IR), C-peptide, and IGF-1 were investigated. We show that hemoglobin-related transcripts (Hbb and 5'-aminolevulinate synthase 2 [Alas2]) increased 46-63% in the running group, while IGF-1-related genes [Igf1 / growth hormone receptor (Ghr)] decreased slightly (7%). There were also moderate to large correlations between Hbb- and IGF-1-related genes in the running group but not in the sedentary group. HOMA-IR, plasma glucose, and insulin changed marginally and non-significantly, but there was a trend toward an increase in plasma-IGF-1 in the running group. In conclusion, seven days of running increased Hbb-related transcripts in three brain regions. Hbb-related transcripts correlated with components of the brain IGF-1 system only in the running group.
Collapse
Affiliation(s)
- Marion Walser
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Region Västra Götaland, Department of Clinical Chemistry, Sahlgrenska University Hospital, Laboratory of Experimental Endocrinology, Bruna Stråket 16, 413 45 , Gothenburg, Sweden.
| | - Lars Karlsson
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Reza Motalleb
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jörgen Isgaard
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Specialist Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - H Georg Kuhn
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Institute for Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johan Svensson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Specialist Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - N David Åberg
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Acute Medicine and Geriatrics, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
4
|
Rathor R, Suryakumar G. Myokines: A central point in managing redox homeostasis and quality of life. Biofactors 2024; 50:885-909. [PMID: 38572958 DOI: 10.1002/biof.2054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/15/2024] [Indexed: 04/05/2024]
Abstract
Redox homeostasis is a crucial phenomenon that is obligatory for maintaining the healthy status of cells. However, the loss of redox homeostasis may lead to numerous diseases that ultimately result in a compromised quality of life. Skeletal muscle is an endocrine organ that secretes hundreds of myokines. Myokines are peptides and cytokines produced and released by muscle fibers. Skeletal muscle secreted myokines act as a robust modulator for regulating cellular metabolism and redox homeostasis which play a prime role in managing and improving metabolic function in multiple organs. Further, the secretory myokines maintain redox homeostasis not only in muscles but also in other organs of the body via stabilizing oxidants and antioxidant levels. Myokines are also engaged in maintaining mitochondrial dynamics as mitochondria is a central point for the generation of reactive oxygen species (ROS). Ergo, myokines also act as a central player in communicating signals to other organs, including the pancreas, gut, liver, bone, adipose tissue, brain, and skin via their autocrine, paracrine, or endocrine effects. The present review provides a comprehensive overview of skeletal muscle-secreted myokines in managing redox homeostasis and quality of life. Additionally, probable strategies will be discussed that provide a solution for a better quality of life.
Collapse
Affiliation(s)
- Richa Rathor
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| | - Geetha Suryakumar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| |
Collapse
|
5
|
Chen W, Siew-Pin JL, Wu Y, Huang N, Teo WP. Identifying exercise and cognitive intervention parameters to optimize executive function in older adults with mild cognitive impairment and dementia: a systematic review and meta-analyses of randomized controlled trials. Eur Rev Aging Phys Act 2024; 21:22. [PMID: 39215230 PMCID: PMC11363393 DOI: 10.1186/s11556-024-00357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Physical exercise is recognized for its beneficial effects on brain health and executive function, particularly through the careful manipulation of key exercise parameters, including type, intensity, and duration. The aim of this systematic review and meta-analysis was to delineate the optimal types, intensities, and durations of exercise that improve cognitive functions in older adults with mild cognitive impairment (MCI) or dementia. A comprehensive search was conducted in Scopus, Web of Science, and PubMed from their inception until December 2023. The methodological quality and publication bias of the included studies were assessed using the PEDro scale and Egger's regression test, respectively. Separate meta-analyses were performed to assess the overall impact of exercise on cognitive assessments and to explore the effects of different exercise types (i.e., aerobic, resistance, dual-task, mind-body, and multi-component exercises) and intensities (i.e., low, moderate, and high) on executive function. Results were presented as standardized mean differences (SMD) and 95% confidence intervals (95% CI). A meta-regression analysis was conducted to examine the correlation between exercise duration and mean effects. In total, 15,087 articles were retrieved from three databases, of which 35 studies were included in our final analyses. The results indicated high overall methodological quality (PEDro score = 8) but a potential for publication bias (t = 2.08, p = 0.045). Meta-analyses revealed that all types of exercise (SMD = 0.691, CI [0.498 to 0.885], p < 0.001) and intensities (SMD = 0.694, CI [0.485 to 0.903], p < 0.001) show significant effects favoring exercise. Notably, dual-task exercises (SMD = 1.136, CI [0.236 to 2.035], p < 0.001) and moderate-intensity exercises (SMD = 0.876, CI [0.533 to 1.219], p < 0.001) exhibited the greatest effect. No significant correlation was observed between exercise duration and SMD (R² = 0.038, p = 0.313). Overall, our meta-analyses support the role of physical exercise in enhancing executive function in older adults with MCI or dementia. It is essential to carefully tailor exercise parameters, particularly type and intensity, to meet the specific needs of older adults with MCI or dementia. Such customization is crucial for optimizing executive function outcomes and improving overall brain health.
Collapse
Affiliation(s)
- Wenxin Chen
- Physical Education College, Hubei University of Arts and Sciences, Hubei, China
| | - Jessie Leuk Siew-Pin
- Physical Education and Sport Science (PESS) Department, National Institute of Education, Nanyang Technological University, Singapore, Singapore
| | - Yuhang Wu
- School of Transportation and Logistics Engineering, Wuhan University of Technology, Wuhan, China
| | - Ning Huang
- School of Public Health, Peking University, Beijing, China
| | - Wei-Peng Teo
- Physical Education and Sport Science (PESS) Department, National Institute of Education, Nanyang Technological University, Singapore, Singapore.
- Science of Learning in Education Centre (SoLEC), National Institute of Education, Nanyang Technological University, 1 Nanyang Walk, Singapore, 637616, Singapore.
| |
Collapse
|
6
|
Noriega-Prieto JA, Maglio LE, Perez-Domper P, Dávila JC, Gutiérrez A, Torres-Alemán I, Fernández de Sevilla D. Bidirectional modulation of synaptic transmission by insulin-like growth factor-I. Front Cell Neurosci 2024; 18:1390663. [PMID: 38910964 PMCID: PMC11193368 DOI: 10.3389/fncel.2024.1390663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/29/2024] [Indexed: 06/25/2024] Open
Abstract
Insulin-like growth factor-I (IGF-I) plays a key role in the modulation of synaptic plasticity and is an essential factor in learning and memory processes. However, during aging, IGF-I levels are decreased, and the effect of this decrease in the induction of synaptic plasticity remains unknown. Here we show that the induction of N-methyl-D-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP) at layer 2/3 pyramidal neurons (PNs) of the mouse barrel cortex is favored or prevented by IGF-I (10 nM) or IGF-I (7 nM), respectively, when IGF-I is applied 1 h before the induction of Hebbian LTP. Analyzing the cellular basis of this bidirectional control of synaptic plasticity, we observed that while 10 nM IGF-I generates LTP (LTPIGF-I) of the post-synaptic potentials (PSPs) by inducing long-term depression (LTD) of the inhibitory post-synaptic currents (IPSCs), 7 nM IGF-I generates LTD of the PSPs (LTDIGF-I) by inducing LTD of the excitatory post-synaptic currents (EPSCs). This bidirectional effect of IGF-I is supported by the observation of IGF-IR immunoreactivity at both excitatory and inhibitory synapses. Therefore, IGF-I controls the induction of Hebbian NMDAR-dependent plasticity depending on its concentration, revealing novel cellular mechanisms of IGF-I on synaptic plasticity and in the learning and memory machinery of the brain.
Collapse
Affiliation(s)
- José Antonio Noriega-Prieto
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Laura Eva Maglio
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Paloma Perez-Domper
- Centro de Investigaciones Biomédicas en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto Cajal (CSIC), Madrid, Spain
| | - José Carlos Dávila
- Centro de Investigaciones Biomédicas en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento Biología Celular, Genética y Fisiología. Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Málaga, Spain
| | - Antonia Gutiérrez
- Centro de Investigaciones Biomédicas en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento Biología Celular, Genética y Fisiología. Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Málaga, Spain
| | - Ignacio Torres-Alemán
- Centro de Investigaciones Biomédicas en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Science Foundation, Bilbao, Spain
| | - David Fernández de Sevilla
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
7
|
Herrero-Labrador R, Fernández-Irigoyen J, Vecino R, González-Arias C, Ausín K, Crespo I, Fernández Acosta FJ, Nieto-Estévez V, Román MJ, Perea G, Torres-Alemán I, Santamaría E, Vicario C. Brain IGF-I regulates LTP, spatial memory, and sexual dimorphic behavior. Life Sci Alliance 2023; 6:e202201691. [PMID: 37463753 PMCID: PMC10355288 DOI: 10.26508/lsa.202201691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Insulin-like growth factor-I (IGF-I) exerts multiple actions, yet the role of IGF-I from different sources is poorly understood. Here, we explored the functional and behavioral consequences of the conditional deletion of Igf-I in the nervous system (Igf-I Δ/Δ), and demonstrated that long-term potentiation was impaired in hippocampal slices. Moreover, Igf-I Δ/Δ mice showed spatial memory deficits in the Morris water maze, and the significant sex-dependent differences displayed by Igf-I Ctrl/Ctrl mice disappeared in Igf-I Δ/Δ mice in the open field and rota-rod tests. Brain Igf-I deletion disorganized the granule cell layer of the dentate gyrus (DG), and it modified the relative expressions of GAD and VGLUT1, which are preferentially localized to inhibitory and excitatory presynaptic terminals. Furthermore, Igf-I deletion altered protein modules involved in receptor trafficking, synaptic proteins, and proteins that functionally interact with estrogen and androgen metabolism. Our findings indicate that brain IGF-I is crucial for long-term potentiation, and that it is involved in the regulation of spatial memory and sexual dimorphic behaviors, possibly by maintaining the granule cell layer structure and the stability of synaptic-related protein modules.
Collapse
Affiliation(s)
- Raquel Herrero-Labrador
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Joaquín Fernández-Irigoyen
- Proteored-ISCIII, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Rebeca Vecino
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Karina Ausín
- Proteored-ISCIII, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Inmaculada Crespo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- CES Cardenal Cisneros, Madrid, Spain
| | | | - Vanesa Nieto-Estévez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - M José Román
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Gertrudis Perea
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Ignacio Torres-Alemán
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Achucarro Basque Center for Neuroscience, and Ikerbasque Foundation for Science, Bilbao, Spain
| | - Enrique Santamaría
- Proteored-ISCIII, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Carlos Vicario
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
8
|
Hjortshoej MH, Aagaard P, Storgaard CD, Juneja H, Lundbye‐Jensen J, Magnusson SP, Couppé C. Hormonal, immune, and oxidative stress responses to blood flow-restricted exercise. Acta Physiol (Oxf) 2023; 239:e14030. [PMID: 37732509 PMCID: PMC10909497 DOI: 10.1111/apha.14030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/12/2023] [Accepted: 07/30/2023] [Indexed: 09/22/2023]
Abstract
INTRODUCTION Heavy-load free-flow resistance exercise (HL-FFRE) is a widely used training modality. Recently, low-load blood-flow restricted resistance exercise (LL-BFRRE) has gained attention in both athletic and clinical settings as an alternative when conventional HL-FFRE is contraindicated or not tolerated. LL-BFRRE has been shown to result in physiological adaptations in muscle and connective tissue that are comparable to those induced by HL-FFRE. The underlying mechanisms remain unclear; however, evidence suggests that LL-BFRRE involves elevated metabolic stress compared to conventional free-flow resistance exercise (FFRE). AIM The aim was to evaluate the initial (<10 min post-exercise), intermediate (10-20 min), and late (>30 min) hormonal, immune, and oxidative stress responses observed following acute sessions of LL-BFRRE compared to FFRE in healthy adults. METHODS A systematic literature search of randomized and non-randomized studies was conducted in PubMed, Embase, Cochrane Central, CINAHL, and SPORTDiscus. The Cochrane Risk of Bias (RoB2, ROBINS-1) and TESTEX were used to evaluate risk of bias and study quality. Data extractions were based on mean change within groups. RESULTS A total of 12525 hits were identified, of which 29 articles were included. LL-BFRRE demonstrated greater acute increases in growth hormone responses when compared to overall FFRE at intermediate (SMD 2.04; 95% CI 0.87, 3.22) and late (SMD 2.64; 95% CI 1.13, 4.16) post-exercise phases. LL-BFRRE also demonstrated greater increase in testosterone responses compared to late LL-FFRE. CONCLUSION These results indicate that LL-BFRRE can induce increased or similar hormone and immune responses compared to LL-FFRE and HL-FFRE along with attenuated oxidative stress responses compared to HL-FFRE.
Collapse
Affiliation(s)
- M. H. Hjortshoej
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Physical and Occupational TherapyBispebjerg and Frederiksberg University HospitalCopenhagenDenmark
- Centre for Health and RehabilitationUniversity College AbsalonSlagelseDenmark
| | - P. Aagaard
- Department of Sports Science and Clinical BiomechanicsUniversity of Southern DenmarkOdenseDenmark
| | - C. D. Storgaard
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Nutrition, Exercise and Sports, Section of Integrative PhysiologyUniversity of CopenhagenCopenhagenDenmark
| | - H. Juneja
- Centre for Health and RehabilitationUniversity College AbsalonSlagelseDenmark
| | - J. Lundbye‐Jensen
- Department of Nutrition, Exercise and Sports, Section of Integrative PhysiologyUniversity of CopenhagenCopenhagenDenmark
| | - S. P. Magnusson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Physical and Occupational TherapyBispebjerg and Frederiksberg University HospitalCopenhagenDenmark
| | - C. Couppé
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Physical and Occupational TherapyBispebjerg and Frederiksberg University HospitalCopenhagenDenmark
| |
Collapse
|
9
|
Alves JM, Smith A, Chow T, Negriff S, Carter S, Xiang AH, Page KA. Prenatal Exposure to Gestational Diabetes Mellitus is Associated with Mental Health Outcomes and Physical Activity has a Modifying Role. RESEARCH SQUARE 2023:rs.3.rs-3290222. [PMID: 37693617 PMCID: PMC10491372 DOI: 10.21203/rs.3.rs-3290222/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Background Studies suggest a link between prenatal gestational diabetes mellitus (GDM) exposure and poor mental health outcomes. We examined associations between prenatal GDM exposure and depressive and anxiety symptoms in children and assessed physical activity as a potential modifier of these associations. Method Seventy children (AgeM(SD): 12(2.0), 56% GDM, 59% female) and their parents completed surveys: Center for Epidemiological Studies Depression Scale for Children (CES-DC), State-Trait Anxiety Inventory for Children (STAIC), Child Behavior Checklist (CBCL), and 3-day physical activity recall (3DPAR). Associations between mental health measures with GDM exposure and interactions between GDM exposure and child moderate-to-vigorous physical activity (MVPA) were assessed using regression. Results GDM-exposed children had higher anxiety (p = 0.03) and internalizing symptoms (CBCL) (p = 0.04) than unexposed children. There was an interaction between GDM exposure and child MVPA on anxiety (p = 0.02), internalizing (p = 0.04) and externalizing symptoms (p = 0.004). In the low MVPA group, GDM exposed children had more depressive (p = 0.03), anxiety (p = 0.003), and internalizing symptoms (p = 0.03) than unexposed children. In the high MVPA group, there were no group differences except with externalizing symptoms (p = 0.04). Conclusion Prenatal GDM is associated with higher anxiety and internalizing symptoms in children. Child MVPA modified the relationship between GDM exposure and mental health outcomes suggesting that physical activity during childhood could mitigate the negative mental health outcomes associated with prenatal GDM exposure.
Collapse
Affiliation(s)
| | | | - Ting Chow
- Kaiser Permanente Southern California
| | | | | | | | | |
Collapse
|
10
|
Hwang D, Kim J, Kyun S, Jang I, Kim T, Park HY, Lim K. Exogenous lactate augments exercise-induced improvement in memory but not in hippocampal neurogenesis. Sci Rep 2023; 13:5838. [PMID: 37037890 PMCID: PMC10086059 DOI: 10.1038/s41598-023-33017-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 04/05/2023] [Indexed: 04/12/2023] Open
Abstract
Adult hippocampal neurogenesis (AHN), the lifelong process of formation of new neurons in the mammalian brain, plays an important role in learning and memory. Exercise is an effective enhancer of AHN; however, the molecular mediators of exercise-induced AHN are unknown. Recently, lactate was considered as an important mediator of exercise-induced AHN. Therefore, we hypothesized that exercise with lactate intake could augment exercise-induced AHN. This study was conducted for 5 weeks with 7-week-old ICR male mice that performed mild-intensity exercise (just below lactate threshold, 55-60%VO2max) with or without oral administration of lactate 5 days/week. Cell proliferation, neuronal differentiation, neurogenesis-relevant factors, reference and retention memory, and spatial working memory were evaluated at the end of the experiment. The results showed that AHN was enhanced by lactate intake, but exercise-induced AHN was not augmented by exercise with lactate intake. Nevertheless, exercise-induced improvement in reference and retention memory was augmented by exercise with lactate intake. And spatial working memory was promoted by the co-treatment, also protein expression of hippocampal FNDC5, BDNF, PGC1α, and MCT2 were elevated by the co-treatment. Therefore, our findings suggest that lactate has a potential to be developed as a novel supplement that improves the positive effects of exercise on the hippocampus and its cognitive function.
Collapse
Affiliation(s)
- Deunsol Hwang
- Laboratory of Exercise and Nutrition, Department of Sports Medicine and Science in Graduate School, Konkuk University, Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul, Republic of Korea
| | - Jisu Kim
- Laboratory of Exercise and Nutrition, Department of Sports Medicine and Science in Graduate School, Konkuk University, Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul, Republic of Korea
| | - Sunghwan Kyun
- Laboratory of Exercise and Nutrition, Department of Sports Medicine and Science in Graduate School, Konkuk University, Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul, Republic of Korea
| | - Inkwon Jang
- Laboratory of Exercise and Nutrition, Department of Sports Medicine and Science in Graduate School, Konkuk University, Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul, Republic of Korea
| | - Taeho Kim
- Laboratory of Exercise and Nutrition, Department of Sports Medicine and Science in Graduate School, Konkuk University, Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul, Republic of Korea
| | - Hun-Young Park
- Laboratory of Exercise and Nutrition, Department of Sports Medicine and Science in Graduate School, Konkuk University, Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul, Republic of Korea
| | - Kiwon Lim
- Laboratory of Exercise and Nutrition, Department of Sports Medicine and Science in Graduate School, Konkuk University, Seoul, Republic of Korea.
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul, Republic of Korea.
- Department of Physical Education, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Alzoubi KH, Halboup AM, Khabour OF, Alomari MA. The Protective Effects of the Combination of Vitamin E and Swimming Exercise on Memory Impairment Induced by Exposure to Waterpipe Smoke. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:304-312. [PMID: 35306997 DOI: 10.2174/1871527321666220318113635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/12/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Waterpipe smoking (WP) exposure involves a negative health impact, including memory deficit, which is attributed to the elevation of oxidative stress. Vitamin E (VitE) in combination with swimming exercise exerts protective effects that prevent memory impairment. In the current study, the modulation of WP-induced memory impairment by the combined effect of VitE and swimming exercise (SE) was investigated. METHODS Animals were exposed to WP one hour/day, five days per week for four weeks. Simultaneously, VitE (100 mg/kg, six days/week for four weeks) was administered via oral gavage, and the rats were made to swim one hour/day, five days/week for four weeks. Changes in memory were evaluated using radial arm water maze (RAWM), and oxidative stress biomarkers were examined in the hippocampus. RESULTS WP exposure induced short-term/long-term memory impairment (p<0.05). This impairment was prevented by a combination of VitE with SE (p<0.05). Additionally, this combination normalized the hippocampal catalase, GPx, and GSH/GSSG ratios that were modulated by WP (p<0.05). The combination further reduced TBARs levels below those of the control group (p<0.05). CONCLUSION WP-induced memory impairments were prevented by the combination of VitE with SE. This could be attributed to preserving the hippocampal oxidative mechanism by combining VitE and SE during WP exposure.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Abdulsalam M Halboup
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, University of Science and Technology, Sana\'a, Yemen
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Jordan
| | - Mahmoud A Alomari
- Department of Rehabilitation Sciences, Division of Physical Therapy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
12
|
Zhang Y, Sun Q, Li H, Wang D, Wang Y, Wang Z. Lower serum insulin-like growth factor 1 concentrations in patients with chronic insomnia disorder. Front Psychiatry 2023; 14:1102642. [PMID: 37151979 PMCID: PMC10160412 DOI: 10.3389/fpsyt.2023.1102642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Objectives Insulin-like growth factor 1 (IGF-1) is a crucial neurotrophin that is produced in the brain and periphery and may play an important role in insomnia and mood disorders. We aimed to analyze its serum concentrations in patients with chronic insomnia disorder (CID). Methods Patients with CID were enrolled in this study and divided into the CID group [Generalized Anxiety Disorder-7 (GAD-7) score < 10] and the CID with anxiety group (GAD-7 score ≥ 10). Age-and sex-matched healthy volunteers were recruited as controls. The Pittsburgh Sleep Quality Index (PSQI) was used to assess sleep quality and the GAD-7 and the Patient Health Questionnaire-9 to assess emotional status. All subjects were monitored via polysomnography, and the serum IGF-1 concentrations in their peripheral blood were detected via enzyme-linked immunosorbent assays. Results We enrolled 65 patients with CID (of whom 35 had anxiety) and 36 controls. The PSQI score and IGF-1 concentration in the CID and CID with anxiety groups were higher than those in the control group. The apparent difference in IGF-1 concentration between the CID and CID with anxiety groups was not statistically significant. The IGF-1 concentration in patients with CID was linearly correlated with the GAD-7 score, PSQI score, and stage 3 non-rapid eye movement (stage N3) time. Conclusion The serum IGF-1 concentration in patients with CID was lower than that of participants without CID, negatively correlated with anxiety score and sleep quality, and positively correlated with stage N3 time.
Collapse
|
13
|
Rai M, Demontis F. Muscle-to-Brain Signaling Via Myokines and Myometabolites. Brain Plast 2022; 8:43-63. [PMID: 36448045 PMCID: PMC9661353 DOI: 10.3233/bpl-210133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle health and function are important determinants of systemic metabolic homeostasis and organism-wide responses, including disease outcome. While it is well known that exercise protects the central nervous system (CNS) from aging and disease, only recently this has been found to depend on the endocrine capacity of skeletal muscle. Here, we review muscle-secreted growth factors and cytokines (myokines), metabolites (myometabolites), and other unconventional signals (e.g. bioactive lipid species, enzymes, and exosomes) that mediate muscle-brain and muscle-retina communication and neuroprotection in response to exercise and associated processes, such as the muscle unfolded protein response and metabolic stress. In addition to impacting proteostasis, neurogenesis, and cognitive functions, muscle-brain signaling influences complex brain-dependent behaviors, such as depression, sleeping patterns, and biosynthesis of neurotransmitters. Moreover, myokine signaling adapts feeding behavior to meet the energy demands of skeletal muscle. Contrary to protective myokines induced by exercise and associated signaling pathways, inactivity and muscle wasting may derange myokine expression and secretion and in turn compromise CNS function. We propose that tailoring muscle-to-CNS signaling by modulating myokines and myometabolites may combat age-related neurodegeneration and brain diseases that are influenced by systemic signals.
Collapse
Affiliation(s)
- Mamta Rai
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
14
|
Hernández-Ramírez S, Salcedo-Tello P, Osorio-Gómez D, Bermúdez-Rattoni F, Pacheco-López G, Ferreira G, Lafenetre P, Guzmán-Ramos KR. Voluntary physical activity improves spatial and recognition memory deficits induced by post-weaning chronic exposure to a high-fat diet. Physiol Behav 2022; 254:113910. [PMID: 35820628 DOI: 10.1016/j.physbeh.2022.113910] [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: 02/08/2022] [Revised: 07/02/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022]
Abstract
Childhood and adolescent exposure to obesogenic environments has contributed to the development of several health disorders, including neurocognitive impairment. Adolescence is a critical neurodevelopmental window highly influenced by environmental factors that affect brain function until adulthood. Post-weaning chronic exposure to a high-fat diet (HFD) adversely affects memory performance; physical activity is one approach to coping with these dysfunctions. Previous studies indicate that voluntary exercise prevents HFD's detrimental effects on memory; however, it remains to evaluate whether it has a remedial/therapeutical effect when introduced after a long-term HFD exposure. This study was conducted on a diet-induced obesity mice model over six months. After three months of HFD exposure (without interrupting the diet) access to voluntary physical activity was provided. HFD produced weight gain, increased adiposity, and impaired glucose tolerance. Voluntary physical exercise ameliorated glucose tolerance and halted weight gain and fat accumulation. Additionally, physical activity mitigated HFD-induced spatial and recognition memory impairments. Our data indicate that voluntary physical exercise starting after several months of periadolescent HFD exposure reverses metabolic and cognitive alterations demonstrating that voluntary exercise, in addition to its known preventive effect, also has a restorative impact on metabolism and cognition dysfunctions associated with obesity.
Collapse
Affiliation(s)
- Susana Hernández-Ramírez
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Av. de las Garzas No. 10, Lerma de Villada, Estado de México, C.P. 52005, Mexico
| | - Pamela Salcedo-Tello
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana (UAM), Unidad Lerma. Av. de las Garzas No. 10, Col. el Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico
| | - Daniel Osorio-Gómez
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México (UNAM). Circuito Exterior, Ciudad Universitaria, 04510 Mexico City
| | - Federico Bermúdez-Rattoni
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México (UNAM). Circuito Exterior, Ciudad Universitaria, 04510 Mexico City
| | - Gustavo Pacheco-López
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana (UAM), Unidad Lerma. Av. de las Garzas No. 10, Col. el Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico
| | - Guillaume Ferreira
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro Laboratory, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Pauline Lafenetre
- Université de Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France
| | - Kioko R Guzmán-Ramos
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana (UAM), Unidad Lerma. Av. de las Garzas No. 10, Col. el Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico.
| |
Collapse
|
15
|
Marks JR, Beatty AE, Husak JF, Schwartz TS, Lailvaux SP. Sprint training interacts with body mass to affect hepatic insulin-like growth factor expression in female green anoles (Anolis carolinensis). Gen Comp Endocrinol 2022; 327:114067. [PMID: 35640679 DOI: 10.1016/j.ygcen.2022.114067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/04/2022]
Abstract
Locomotor performance is a key predictor of fitness in many animal species. As such, locomotion integrates the output of a number of morphological, physiological, and molecular levels of organization, yet relatively little is known regarding the major molecular pathways that bolster locomotor performance. One potentially relevant pathway is the insulin and insulin-like signaling (IIS) network, a significant regulator of physiological processes such as reproduction, growth, and metabolism. Two primary hormones of this network, insulin-like growth factor 1 (IGF1) and insulin-like growth factor 2 (IGF2) are important mediators of these processes and, consequently, of life-history strategies. We sprint-trained green anole (Anolis carolinensis) females to test the responsiveness of IGF1 and IGF2 hepatic gene expression to exercise training. We also tested how sprint training would affect glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and eukaryotic elongation factor 2 (EEF2). The former is a crucial enzyme for glycolytic function in a cell, and the latter is necessary for protein synthesis. Resistance exercise forces animals to increase investment of resources towards skeletal muscle growth. Because IGF1 and IGF2 are important hormones for growth, and GAPDH and EEF2 are crucial for proper cellular function, we hypothesized that these four genes would be affected by sprint training. We found that sprint training affects IGF and EEF2 expression, such that larger sprint-trained lizards express hepatic IGF1, IGF2, and EEF2 to a lesser extent than similarly sized untrained lizards. These results demonstrate that the IIS, and pathways connected to it, can react in a size-dependent manner and are implicated in the exercise response in reptiles.
Collapse
Affiliation(s)
- Jamie R Marks
- Department of Biological Sciences, University of New Orleans, 2000 Lakeshore Dr., New Orleans, LA 70148, USA.
| | - Abby E Beatty
- Department of Biological Sciences, Auburn University, 101 Rouse Life Sciences Bldg, Auburn, AL 36849, USA
| | - Jerry F Husak
- Department of Biology, University of St. Thomas, St Paul, MN 55105, USA
| | - Tonia S Schwartz
- Department of Biological Sciences, Auburn University, 101 Rouse Life Sciences Bldg, Auburn, AL 36849, USA
| | - Simon P Lailvaux
- Department of Biological Sciences, University of New Orleans, 2000 Lakeshore Dr., New Orleans, LA 70148, USA
| |
Collapse
|
16
|
Rowe RK, Griesbach GS. Immune-endocrine interactions in the pathophysiology of sleep-wake disturbances following traumatic brain injury: A narrative review. Brain Res Bull 2022; 185:117-128. [DOI: 10.1016/j.brainresbull.2022.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 12/16/2022]
|
17
|
Targeting Insulin-Like Growth Factor-I in Management of Neurological Disorders. Neurotox Res 2022; 40:874-883. [PMID: 35476315 DOI: 10.1007/s12640-022-00513-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/29/2022] [Accepted: 04/15/2022] [Indexed: 12/13/2022]
Abstract
The degradation of neurons marks as the pathological reason for onset of most of the neurological diseases although the functional deficiencies and symptoms may vary. Insulin-like growth factor-I (IGF-I) boosts regeneration of both motor and sensory neurons and thus presents as a potential treatment in management of neurological disorders. IGF-I is a pleiotropic agent which stimulates the survival and outgrowth of neurons accompanied by their motility as well as myelination by glial cells. This hormone has been found to possess neuroprotective properties which is in association with its antioxidant and mitochondrial protection activity. Studying and exploring the signaling pathways which mediate pleotropic responses intracellularly have elucidated significant therapeutic approach in treatment and management of neurological disorders by IGF-I. The current review highlights the role of IGF-I in management of major neurological disorders such as depression, Parkinson's disease, and Alzheimer's disease and also covers the mechanisms involved in the process.
Collapse
|
18
|
Koehl M, Ladevèze E, Montcouquiol M, Abrous DN. Vangl2, a Core Component of the WNT/PCP Pathway, Regulates Adult Hippocampal Neurogenesis and Age-Related Decline in Cognitive Flexibility. Front Aging Neurosci 2022; 14:844255. [PMID: 35370613 PMCID: PMC8965557 DOI: 10.3389/fnagi.2022.844255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Decline in episodic memory is one of the hallmarks of aging and represents one of the most important health problems facing Western societies. A key structure in episodic memory is the hippocampal formation and the dentate gyrus in particular, as the continuous production of new dentate granule neurons in this brain region was found to play a crucial role in memory and age-related decline in memory. As such, understanding the molecular processes that regulate the relationship between adult neurogenesis and aging of memory function holds great therapeutic potential. Recently, we found that Vang-Gogh like 2 (Vangl2), a core component of the Planar Cell Polarity (PCP) signaling pathway, is enriched in the dentate gyrus of adult mice. In this context, we sought to evaluate the involvement of this member of the Wnt/PCP pathway in both adult neurogenesis and memory abilities in adult and middle-aged mice. Using a heterozygous mouse model carrying a dominant-negative mutation in the Vangl2 gene, called Looptail (Vangl2Lp), we show that alteration in Vangl2 expression decreases the survival of adult-born granule cells and advances the onset of a decrease in cognitive flexibility. The inability of mutant mice to erase old irrelevant information to the benefit of new relevant ones highlights a key role of Vangl2 in interference-based forgetting. Taken together, our findings show that Vangl2 activity may constitute an interesting target to prevent age-related decline in hippocampal plasticity and memory.
Collapse
Affiliation(s)
- Muriel Koehl
- Univ. Bordeaux, INSERM, Magendie, U1215, Neurogenesis and Pathophysiology group, Bordeaux, France
- *Correspondence: Muriel Koehl
| | - Elodie Ladevèze
- Univ. Bordeaux, INSERM, Magendie, U1215, Neurogenesis and Pathophysiology group, Bordeaux, France
| | - Mireille Montcouquiol
- Univ. Bordeaux, INSERM, Magendie, U1215, Planar Polarity and Plasticity Group, Bordeaux, France
| | - Djoher Nora Abrous
- Univ. Bordeaux, INSERM, Magendie, U1215, Neurogenesis and Pathophysiology group, Bordeaux, France
| |
Collapse
|
19
|
Hugues N, Pin-Barre C, Pellegrino C, Rivera C, Berton E, Laurin J. Time-Dependent Cortical Plasticity during Moderate-Intensity Continuous Training Versus High-Intensity Interval Training in Rats. Cereb Cortex 2022; 32:3829-3847. [PMID: 35029628 DOI: 10.1093/cercor/bhab451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/14/2022] Open
Abstract
The temporal pattern of cortical plasticity induced by high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) is required to clarify their relative benefits to prevent neurological disorders. The purpose of this study is to define the time-dependent effects of work-matched HIIT and MICT on cortical plasticity, endurance, and sensorimotor performances over an 8-week training period in healthy rats. Adult healthy rats performed incremental exercise tests and sensorimotor tests before and at 2, 4, and 8 weeks of training. In parallel, cortical markers related to neurotrophic, angiogenic, and metabolic activities were assessed. Results indicate that HIIT induced an early and superior endurance improvement compared to MICT. We found significant enhancement of speed associated with lactate threshold (SLT) and maximal speed (Smax) in HIIT animals. MICT promoted an early increase in brain-derived neurotrophic factor and angiogenic/metabolic markers but showed less influence at 8 weeks. HIIT upregulated the insulin-like growth factor-1 (IGF-1) as well as neurotrophic, metabolic/angiogenic markers at 2 and 8 weeks and downregulated the neuronal K-Cl cotransporter KCC2 that regulates GABAA-mediated transmission. HIIT and MICT are effective in a time-dependent manner suggesting a complementary effect that might be useful in physical exercise guidelines for maintaining brain health.
Collapse
Affiliation(s)
- Nicolas Hugues
- Aix-Marseille Univ, INSERM, INMED, Marseille, France
- Aix-Marseille Univ, CNRS, ISM, Marseille, France
| | | | | | | | - Eric Berton
- Aix-Marseille Univ, CNRS, ISM, Marseille, France
| | - Jérôme Laurin
- Aix-Marseille Univ, INSERM, INMED, Marseille, France
| |
Collapse
|
20
|
Hilal S, Brayne C. Epidemiologic Trends, Social Determinants, and Brain Health: The Role of Life Course Inequalities. Stroke 2022; 53:437-443. [PMID: 35000426 DOI: 10.1161/strokeaha.121.032609] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Brain health as expressed in our mental health and occurrence of specific disorders such as dementia and stroke is vitally important to quality of life, functional independence, and risk of institutionalization. Maintaining brain health is, therefore, a societal imperative, and public health challenge, from prevention of acquisition of brain disorders, through protection and risk reduction to supporting those with such disorders through effective societal and system approaches. To identify possible mechanisms that explain the differential effect of potentially modifiable risk factors, and factors that may mitigate risk, a life course approach is needed. This is key to understanding how poor health can accumulate from the earliest life stages. It also allows us to integrate and investigate key material, behavioral, and psychological factors that generate health inequalities within and across communities and societies. This review provides a narrative on how brain health is intimately linked to wider health determinants, thus importance for clinicians and societies alike. There is compelling evidence accumulated from research over decades that socioeconomic status, higher education, and healthy lifestyle extend life and compress major morbidities into later life. Brain health is part of this, but collective action has been limited, partly because of the separation of disciplines and focus on highly reductionist approaches in that clinicians and associated research have focused more on mitigation and early detection of specific diseases. However, clinicians could be part of the drive for better brain health for all society to support life courses that have more protection and less risk. There is evidence of change in such risks for conditions such as stroke and dementia across generations. The evidence points to the importance of starting with parental health and life course inequalities as a central focus.
Collapse
Affiliation(s)
- Saima Hilal
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System (S.H.).,Memory Aging and Cognition Center, National University Health System, Singapore (S.H.).,Department of Pharmacology, National University of Singapore (S.H.)
| | - Carol Brayne
- Department of Public Health and Primary Care, Cambridge Institute of Public Health, School of Clinical Medicine, University of Cambridge, United Kingdom (C.B.)
| |
Collapse
|
21
|
Tomiga Y, Sakai K, Ra SG, Kusano M, Ito A, Uehara Y, Takahashi H, Kawanaka K, Soejima H, Higaki Y. Short-term running exercise alters DNA methylation patterns in neuronal nitric oxide synthase and brain-derived neurotrophic factor genes in the mouse hippocampus and reduces anxiety-like behaviors. FASEB J 2021; 35:e21767. [PMID: 34325488 DOI: 10.1096/fj.202100630r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/14/2021] [Accepted: 06/14/2021] [Indexed: 02/04/2023]
Abstract
Running exercise has beneficial effects on brain health. However, the effects of relatively short-term running exercise (STEx) on behavior, and its underlying signaling pathways, are poorly understood. In this study, we evaluated the possibility that the regulation by STEx of brain-derived neurotrophic factor (BDNF) and neuronal nitric oxide synthase (nNOS, encoded by NOS1), which are important molecules for anxiety regulation, might involve mechanisms of epigenetic modification, such as DNA methylation. C57BL/6J male mice were divided into sedentary (SED, n = 12) and STEx (EX, n = 15) groups; STEx was conducted with the mice for a duration of 11 days. STEx reduced anxiety-like behaviors, and STEx reduced Nos1α and increased Bdnf exon I and IV mRNA levels in the hippocampus. Interestingly, behavioral parameters were associated with Bdnf exon I and IV and Nos1α mRNA levels in the ventral, but not dorsal, hippocampal region. However, STEx had no effect on peroxisome proliferator-activated receptor-γ coactivator 1α (Pgc-1α) or fibronectin type III domain-containing 5 (Fndc5) mRNA levels, which are relatively long-term exercise-induced upstream regulators of BDNF. In parallel with gene expression changes, we found, for the first time, that STEx downregulated Bdnf promoter IV and upregulated Nos1 DNA methylation levels in the hippocampus, and these patterns were partially different between the dorsal and ventral regions. These findings suggest that the beneficial effects of running exercise on mood regulation may be controlled by alterations in epigenetic mechanisms, especially in the ventral hippocampus. These effects occur even after a relatively short-term period of exercise.
Collapse
Affiliation(s)
- Yuki Tomiga
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.,Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Kazuya Sakai
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Song-Gyu Ra
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.,Institute of Liberal Arts and Sciences, Tokushima University, Tokushima, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Masaki Kusano
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Ai Ito
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
| | - Yoshinari Uehara
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Hirokazu Takahashi
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan.,Liver Center, Saga University Hospital, Saga, Japan
| | - Kentaro Kawanaka
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Hidenobu Soejima
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Yasuki Higaki
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
22
|
The interplay of neurovasculature and adult hippocampal neurogenesis. Neurosci Lett 2021; 760:136071. [PMID: 34147540 DOI: 10.1016/j.neulet.2021.136071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/06/2021] [Accepted: 06/15/2021] [Indexed: 01/14/2023]
Abstract
The subgranular zone of the dentate gyrus provides a local microenvironment (niche) for neural stem cells. In the adult brain, it has been established that the vascular compartment of such niches has a significant role in regulating adult hippocampal neurogenesis. More recently, evidence showed that neurovascular coupling, the relationship between blood flow and neuronal activity, also regulates hippocampal neurogenesis. Here, we review the most recent articles on addressing the intricate relationship between neurovasculature and adult hippocampal neurogenesis and a novel pathway where functional hyperemia enhances hippocampal neurogenesis. In the end, we have further reviewed recent research showing that impaired neurovascular coupling may cause declined neurogenesis and contribute to brain damage in neurodegenerative diseases.
Collapse
|
23
|
Maglio LE, Noriega-Prieto JA, Maroto IB, Martin-Cortecero J, Muñoz-Callejas A, Callejo-Móstoles M, Fernández de Sevilla D. IGF-1 facilitates extinction of conditioned fear. eLife 2021; 10:e67267. [PMID: 33792539 PMCID: PMC8043742 DOI: 10.7554/elife.67267] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/31/2021] [Indexed: 11/26/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) plays a key role in synaptic plasticity, spatial learning, and anxiety-like behavioral processes. While IGF-1 regulates neuronal firing and synaptic transmission in many areas of the central nervous system, its signaling and consequences on excitability, synaptic plasticity, and animal behavior dependent on the prefrontal cortex remain unexplored. Here, we show that IGF-1 induces a long-lasting depression of the medium and slow post-spike afterhyperpolarization (mAHP and sAHP), increasing the excitability of layer 5 pyramidal neurons of the rat infralimbic cortex. Besides, IGF-1 mediates a presynaptic long-term depression of both inhibitory and excitatory synaptic transmission in these neurons. The net effect of this IGF-1-mediated synaptic plasticity is a long-term potentiation of the postsynaptic potentials. Moreover, we demonstrate that IGF-1 favors the fear extinction memory. These results show novel functional consequences of IGF-1 signaling, revealing IGF-1 as a key element in the control of the fear extinction memory.
Collapse
Affiliation(s)
- Laura E Maglio
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de MadridMadridSpain
- Departamento de Ciencias Médicas Básicas (Fisiología) and Instituto de Tecnologías Biomédicas (ITB), Universidad de La LagunaTenerifeSpain
| | - José A Noriega-Prieto
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de MadridMadridSpain
- Department of Neuroscience, University of MinnesotaMinneapolisUnited States
| | - Irene B Maroto
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de MadridMadridSpain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Universitario de Investigación Neuroquímica (IUIN), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Departamento de Bioquímica y Biología Molecular, Facultad de Química, Universidad Complutense de MadridMadridSpain
| | - Jesús Martin-Cortecero
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de MadridMadridSpain
- Institute of Physiology and Pathophysiology, Medical Biophysic, Heidelberg UniversityHeidelbergGermany
| | - Antonio Muñoz-Callejas
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de MadridMadridSpain
| | - Marta Callejo-Móstoles
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de MadridMadridSpain
| | - David Fernández de Sevilla
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de MadridMadridSpain
| |
Collapse
|
24
|
Karlsson L, González-Alvarado MN, Motalleb R, Wang Y, Wang Y, Börjesson M, Zhu C, Kuhn HG. Constitutive PGC-1α Overexpression in Skeletal Muscle Does Not Contribute to Exercise-Induced Neurogenesis. Mol Neurobiol 2021; 58:1465-1481. [PMID: 33200398 PMCID: PMC7932943 DOI: 10.1007/s12035-020-02189-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/27/2020] [Indexed: 01/04/2023]
Abstract
Physical exercise can improve age-dependent decline in cognition, which in rodent is partly mediated by restoration of an age-dependent decline in neurogenesis. Exercise-inducible myokines in the circulation present a link in muscle-brain crosstalk. The transcription factor PGC-1α regulates the release of such myokines with neurotrophic properties into the circulation. We study how chronic muscular overexpression of PGC-1α could contribute to exercise-induced effects on hippocampal neurogenesis and if this effect could be enhanced in a running wheel paradigm. We used 3- and 11-month-old transgenic mice with overexpression of PGC-1α under the control of muscle creatinine kinase promoter (MCK-PGC-1α), which have a constitutively developed endurance muscle phenotype. Wild-type and MCK-PGC-1α mice were single housed with free access to running wheels. Four weeks of running in female animals increased the levels of newborn cells, immature neurons, and, for young animals, new mature neurons, compared to sedentary controls. However, no difference in these parameters was observed between wild-type and transgenic mice under sedentary or running conditions. Multiplex analysis of serum cytokines, chemokines, and myokines suggested several differences in serum protein concentrations between genotypes with musclin found to be significantly upregulated 4-fold in male MCK-PGC-1α animals. We conclude that constitutive muscular overexpression of PGC-1α, despite systemic changes and difference in serum composition, does not translate into exercise-induced effects on hippocampal neurogenesis, independent of the age of the animal. This suggests that chronic activation of PGC-1α in skeletal muscle is by itself not sufficient to mimic exercise-induced effects or to prevent decline of neurogenesis in aging.
Collapse
Affiliation(s)
- Lars Karlsson
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
- The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Region of Western Sweden, Gothenburg, Sweden.
| | - María Nazareth González-Alvarado
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Reza Motalleb
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yafeng Wang
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yong Wang
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mats Börjesson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy and Center for Health and Performance, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital/Östra, Region of Western Sweden, Gothenburg, Sweden
| | - Changlian Zhu
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hans-Georg Kuhn
- Center for Brain Repair and Rehabilitation, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
25
|
Butruille L, Vancamp P, Demeneix BA, Remaud S. Thyroid hormone regulation of adult neural stem cell fate: A comparative analysis between rodents and primates. VITAMINS AND HORMONES 2021; 116:133-192. [PMID: 33752817 DOI: 10.1016/bs.vh.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thyroid hormone (TH) signaling, a highly conserved pathway across vertebrates, is crucial for brain development and function throughout life. In the adult mammalian brain, including that of humans, multipotent neural stem cells (NSCs) proliferate and generate neuronal and glial progenitors. The role of TH has been intensively investigated in the two main neurogenic niches of the adult mouse brain, the subventricular and the subgranular zone. A key finding is that T3, the biologically active form of THs, promotes NSC commitment toward a neuronal fate. In this review, we first discuss the roles of THs in the regulation of adult rodent neurogenesis, as well as how it relates to functional behavior, notably olfaction and cognition. Most research uncovering these roles of TH in adult neurogenesis was conducted in rodents, whose genetic background, brain structure and rate of neurogenesis are considerably different from that of humans. To bridge the phylogenetic gap, we also explore the similarities and divergences of TH-dependent adult neurogenesis in non-human primate models. Lastly, we examine how photoperiodic length changes TH homeostasis, and how that might affect adult neurogenesis in seasonal species to increase fitness. Several aspects by which TH acts on adult NSCs seem to be conserved among mammals, while we only start to uncover the molecular pathways, as well as how other in- and extrinsic factors are intertwined. A multispecies approach delivering more insights in the matter will pave the way for novel NSC-based therapies to combat neurological disorders.
Collapse
Affiliation(s)
- Lucile Butruille
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Pieter Vancamp
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France.
| |
Collapse
|
26
|
Taniguchi E, Tashiro A, Hattori A, Furuse M, Yasuo S. Photoperiodic changes in hippocampal neurogenesis and plasma metabolomic profiles in relation to depression-like behavior in mice. Behav Brain Res 2021; 403:113136. [PMID: 33482168 DOI: 10.1016/j.bbr.2021.113136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 01/01/2023]
Abstract
Photoperiod alters affective behaviors and brain neuroplasticity in several mammalian species. We addressed whether neurogenesis and signaling pathways of insulin-like growth factor-I (IGF-I), a key modulator of neuroplasticity, are regulated by photoperiod in C57BL/6 J mice, a putative model of seasonal affective disorder. We also examined the effects of photoperiod on plasma metabolomic profiles in relation to depression-like behavior to understand a possible linkage between peripheral metabolism and behavior. Mice that were maintained under long-day conditions (LD) exhibited a higher number of 5-bromo-2'-deoxyuridine-positive cells and higher levels of astrocyte marker in the dentate gyrus of the hippocampus compared to that of mice under short-day conditions (SD). Plasma IGF-I levels and levels/expression of IGF-I signaling molecules in the hippocampus (Brn-4, NeuroD1, and phospho-Akt) involved in neuronal proliferation and differentiation were higher in the mice under LD. Metabolome analysis using plasma of the mice under LD and SD identified several metabolites that were highly correlated with immobility in the forced swim test, a depression-like behavior. Negative correlations with behavior occurred in the levels of 23 metabolites, including metabolites related to neurogenesis and antidepressant-like effects of exercise, metabolites in the biosynthesis of arginine, and the occurrence of branched chain amino acids. Three metabolites had positive correlations with the behavior, including guanidinosuccinic acid, a neurotoxin. Taken together, photoperiodic responses of neurogenesis and neuro-glial organization in the hippocampus may be involved in photoperiodic alteration of depression-like behavior, mediated through multiple pathways, including IGF-I and peripheral metabolites.
Collapse
Affiliation(s)
- Emi Taniguchi
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Ayako Tashiro
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Ayumi Hattori
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Mitsuhiro Furuse
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shinobu Yasuo
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
27
|
Effects of Physical Exercise on Neuroplasticity and Brain Function: A Systematic Review in Human and Animal Studies. Neural Plast 2021; 2020:8856621. [PMID: 33414823 PMCID: PMC7752270 DOI: 10.1155/2020/8856621] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/02/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Background Physical exercise (PE) has been associated with increase neuroplasticity, neurotrophic factors, and improvements in brain function. Objective To evaluate the effects of different PE protocols on neuroplasticity components and brain function in a human and animal model. Methods We conducted a systematic review process from November 2019 to January 2020 of the following databases: PubMed, ScienceDirect, SciELO, LILACS, and Scopus. A keyword combination referring to PE and neuroplasticity was included as part of a more thorough search process. From an initial number of 20,782 original articles, after reading the titles and abstracts, twenty-one original articles were included. Two investigators evaluated the abstract, the data of the study, the design, the sample size, the participant characteristics, and the PE protocol. Results PE increases neuroplasticity via neurotrophic factors (BDNF, GDNF, and NGF) and receptor (TrkB and P75NTR) production providing improvements in neuroplasticity, and cognitive function (learning and memory) in human and animal models. Conclusion PE was effective for increasing the production of neurotrophic factors, cell growth, and proliferation, as well as for improving brain functionality.
Collapse
|
28
|
Television Viewing and Cognitive Dysfunction of Korean Older Adults. Healthcare (Basel) 2020; 8:healthcare8040547. [PMID: 33321807 PMCID: PMC7763643 DOI: 10.3390/healthcare8040547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022] Open
Abstract
This study examined the association between television (TV) viewing and cognitive dysfunction in elderly Koreans. Among participants of the 2014 National Survey of Older Koreans, 9644 were considered in this study. To better identify the association between two factors, propensity score (PS) matching with exact method was used. Finally, 168 viewers and non-viewers each were selected based on estimated PS on key variables and eliminating double matches. Multivariate logistic regression analysis was performed when controlling for possible covariates. Viewers were more likely to have cognitive dysfunction than non-viewers, with significant differences in most covariates. After correcting confounding effects of these covariates with PS matching, TV viewing was found to be a significant risk factor of cognitive dysfunction, along with absence of diagnosed hypertension and non-participation in physical leisure activities. TV viewing might be associated with increased risk of cognitive dysfunction in later life. Appropriate education and strategies to minimize TV viewing among older adults should be established to contribute to attenuating cognitive aging. More interventional studies can help older adults, caregivers, and healthcare professionals explore the cognitively beneficial alternatives to TV use considering the impact of socioeconomic factors of selecting TV viewing as a preferred leisure activity.
Collapse
|
29
|
Treadmill Exercise Buffers Behavioral Alterations Related to Ethanol Binge-Drinking in Adolescent Mice. Brain Sci 2020; 10:brainsci10090576. [PMID: 32825478 PMCID: PMC7563508 DOI: 10.3390/brainsci10090576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/19/2020] [Indexed: 01/07/2023] Open
Abstract
The binge-drinking pattern of EtOH consumption, which is frequently observed in adolescents, is known to induce several neurobehavioral alterations, but protection strategies against these impairments remain scarcely explored. We aimed to study the protective role of treadmill physical exercise on the deficits caused after repeated cycles of binge-like EtOH exposure in the cognition, motivation, exploration, and emotion of C57BL/6J mice from adolescence to adulthood. Animals were divided into four groups: control group, exercised group, EtOH group, and exercised + EtOH group (20% in tap water). The exercise was performed for 20 min, 5 days/week at 20 cm/s. Then, animals were submitted to several behavioral tasks. Compared to binge-drinking mice, the exercised + EtOH group exhibited diminished anxiolytic-related behaviors in the elevated plus-maze, enhanced exploratory activity in the open field, reduced preference for alcohol odor when another rewarding stimulus was present (social stimulus) and lower latency to start self-cleaning behaviors in the sucrose splash test. In contrast, other measurements such as habituation learning and working memory were not improved by exercise. Besides, exercise was not able to reduce alcohol consumption across the weeks. In conclusion, physical activity during adolescence and early adulthood could buffer certain neurobehavioral alterations associated with binge-drinking, despite not reducing the quantity of consumed alcohol.
Collapse
|
30
|
Bettio LEB, Thacker JS, Rodgers SP, Brocardo PS, Christie BR, Gil-Mohapel J. Interplay between hormones and exercise on hippocampal plasticity across the lifespan. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165821. [PMID: 32376385 DOI: 10.1016/j.bbadis.2020.165821] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/19/2020] [Accepted: 04/25/2020] [Indexed: 12/15/2022]
Abstract
The hippocampus is a brain structure known to play a central role in cognitive function (namely learning and memory) as well as mood regulation and affective behaviors due in part to its ability to undergo structural and functional changes in response to intrinsic and extrinsic stimuli. While structural changes are achieved through modulation of hippocampal neurogenesis as well as alterations in dendritic morphology and spine remodeling, functional (i.e., synaptic) changes can be noted through the strengthening (i.e., long-term potentiation) or weakening (i.e., long-term depression) of the synapses. While age, hormone homeostasis, and levels of physical activity are some of the factors known to module these forms of hippocampal plasticity, the exact mechanisms through which these factors interact with each other at a given moment in time are not completely understood. It is well known that hormonal levels vary throughout the lifespan of an individual and it is also known that physical exercise can impact hormonal homeostasis. Thus, it is reasonable to speculate that hormone modulation might be one of the various mechanisms through which physical exercise differently impacts hippocampal plasticity throughout distinct periods of an individual's life. The present review summarizes the potential relationship between physical exercise and different types of hormones (namely sex, metabolic, and stress hormones) and how this relationship may mediate the effects of physical activity during three distinct life periods, adolescence, adulthood, and senescence. Overall, the vast majority of studies support a beneficial role of exercise in maintaining hippocampal hormonal levels and consequently, hippocampal plasticity, cognition, and mood regulation.
Collapse
Affiliation(s)
- Luis E B Bettio
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Jonathan S Thacker
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Shaefali P Rodgers
- Developmental, Cognitive & Behavioral Neuroscience Program, Department of Psychology, Texas Institute for Measurement, Evaluation, and Statistics, University of Houston, TX, USA
| | - Patricia S Brocardo
- Department of Morphological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Brian R Christie
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada; Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada; Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC, Canada.
| |
Collapse
|
31
|
The Effect of a Multicomponent Dual-Task Exercise on Cortical Thickness in Older Adults with Cognitive Decline: A Randomized Controlled Trial. J Clin Med 2020; 9:jcm9051312. [PMID: 32370159 PMCID: PMC7290566 DOI: 10.3390/jcm9051312] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 02/03/2023] Open
Abstract
The aim of this study was to examine cortical thickness changes associated with a multicomponent exercise intervention combining physical exercise and cognitive training in older adults with cognitive decline. This study involved a secondary analysis of neuroimaging data from a randomized controlled trial with 280 older adults having cognitive decline who were randomly assigned to either a multicomponent exercise group (n = 140) that attended weekly 90-minute exercise and cognitive training sessions or a health education control group (n = 140). The cortical thickness and cognitive performance were assessed at the baseline and at trial completion (10 months). The cortical thickness in the frontal and temporal regions was determined using FreeSurfer software. Cognitive performance was evaluated using the Gerontology-Functional Assessment Tool (NCGG-FAT). The cortical thickness significantly increased in the middle temporal (p < 0.001) and temporal pole (p < 0.001) in the multicomponent exercise group compared with the control group. Cortical thickness changes were significantly associated with change in trail making test (TMT)-A, TMT-B, and story memory after a 10-month multicomponent exercise intervention. This study suggests that multicomponent exercise programs combining physical exercise and cognitive training have important implications for brain health, especially in providing protection from age-related cortical thinning.
Collapse
|
32
|
Niu X, Zhao Y, Yang N, Zhao X, Zhang W, Bai X, Li A, Yang W, Lu L. Proteasome activation by insulin-like growth factor-1/nuclear factor erythroid 2-related factor 2 signaling promotes exercise-induced neurogenesis. Stem Cells 2020; 38:246-260. [PMID: 31648402 DOI: 10.1002/stem.3102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/11/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022]
Abstract
Physical exercise-induced enhancement of learning and memory and alleviation of age-related cognitive decline in humans have been widely acknowledged. However, the mechanistic relationship between exercise and cognitive improvement remains largely unknown. In this study, we found that exercise-elicited cognitive benefits were accompanied by adaptive hippocampal proteasome activation. Voluntary wheel running increased hippocampal proteasome activity in adult and middle-aged mice, contributing to an acceleration of neurogenesis that could be reversed by intrahippocampal injection of the proteasome inhibitor MG132. We further found that increased levels of insulin-like growth factor-1 (IGF-1) in both serum and hippocampus may be essential for exercise-induced proteasome activation. Our in vitro study demonstrated that IGF-1 stimulated proteasome activity in cultured adult neural progenitor cells (NPCs) by promoting nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), followed by elevated expressions of proteasome subunits such as PSMB5. In contrast, pretreating adult mice with the selective IGF-1R inhibitor picropodophyllin diminished exercise-induced neurogenesis, concurrent with reduced Nrf2 nuclear translocation and proteasome activity. Likewise, lowering Nrf2 expression by RNA interference with bilateral intrahippocampal injections of recombinant adeno-associated viral particles significantly suppressed exercise-induced proteasome activation and attenuated cognitive function. Collectively, our work demonstrates that proteasome activation in hippocampus through IGF-1/Nrf2 signaling is a key adaptive mechanism underlying exercise-related neurogenesis, which may serve as a potential targetable pathway in neurodegeneration.
Collapse
Affiliation(s)
- Xiaojie Niu
- Department of Anatomy, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yunhe Zhao
- Department of Anatomy, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Na Yang
- Department of Anatomy, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xuechun Zhao
- Department of Anatomy, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Wei Zhang
- Department of Anatomy, Shanxi Medical University, Taiyuan, People's Republic of China
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Jinan University, Guangzhou, People's Republic of China
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Jinan University, Guangzhou, People's Republic of China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, People's Republic of China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, People's Republic of China
| | - Li Lu
- Department of Anatomy, Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
33
|
Cicvaric A, Sachernegg HM, Stojanovic T, Symmank D, Smani T, Moeslinger T, Uhrin P, Monje FJ. Podoplanin Gene Disruption in Mice Promotes in vivo Neural Progenitor Cells Proliferation, Selectively Impairs Dentate Gyrus Synaptic Depression and Induces Anxiety-Like Behaviors. Front Cell Neurosci 2020; 13:561. [PMID: 32009902 PMCID: PMC6974453 DOI: 10.3389/fncel.2019.00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Podoplanin (Pdpn), a brain-tumor-related glycoprotein identified in humans and animals, is endogenously expressed in several organs critical for life support such as kidney, lung, heart and brain. In the brain, Pdpn has been identified in proliferative nestin-positive adult neural progenitor cells and in neurons of the neurogenic hippocampal dentate gyrus (DG), a structure associated to anxiety, critical for learning and memory functions and severely damaged in people with Alzheimer's Disease (AD). The in vivo role of Pdpn in adult neurogenesis and anxiety-like behavior remained however unexplored. Using mice with disrupted Pdpn gene as a model organism and applying combined behavioral, molecular biological and electrophysiological assays, we here show that the absence of Pdpn selectively impairs long-term synaptic depression in the neurogenic DG without affecting the CA3-Schaffer's collateral-CA1 synapses. Pdpn deletion also enhanced the proliferative capacity of DG neural progenitor cells and diminished survival of differentiated neuronal cells in vitro. In addition, mice with podoplanin gene disruption showed increased anxiety-like behaviors in experimentally validated behavioral tests as compared to wild type littermate controls. Together, these findings broaden our knowledge on the molecular mechanisms influencing hippocampal synaptic plasticity and neurogenesis in vivo and reveal Pdpn as a novel molecular target for future studies addressing general anxiety disorder and synaptic depression-related memory dysfunctions.
Collapse
Affiliation(s)
- Ana Cicvaric
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannah M. Sachernegg
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Dörte Symmank
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville (IBiS)/University of Seville/CIBERCV, Seville, Spain
| | - Thomas Moeslinger
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
34
|
Potential exerkines for physical exercise-elicited pro-cognitive effects: Insight from clinical and animal research. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 147:361-395. [PMID: 31607361 DOI: 10.1016/bs.irn.2019.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A sedentary lifestyle is now known as a critical risk factor for accelerated aging-related neurodegenerative disorders. In contract, having regular physical exercise has opposite effects. Clinical findings have suggested that physical exercise can promote brain plasticity, particularly the hippocampus and the prefrontal cortex, that are important for learning and memory and mood regulations. However, the underlying mechanisms are still unclear. Animal studies reveal that the effects of physical exercise on promoting neuroplasticity could be mediated by different exerkines derived from the peripheral system and the brain itself. This book chapter summarizes the recent evidence from clinical and pre-clinical studies showing the emerging mediators for exercise-promoted brain health, including myokines secreted from skeletal muscles, adipokines from adipose tissues, and other factors secreted from the bone and liver.
Collapse
|
35
|
Marston KJ, Brown BM, Rainey-Smith SR, Peiffer JJ. Resistance Exercise-Induced Responses in Physiological Factors Linked with Cognitive Health. J Alzheimers Dis 2019; 68:39-64. [DOI: 10.3233/jad-181079] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Kieran J. Marston
- Department of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
| | - Belinda M. Brown
- Department of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Australian Alzheimer’s Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Stephanie R. Rainey-Smith
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Australian Alzheimer’s Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Jeremiah J. Peiffer
- Department of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
- Ageing, Cognition and Exercise (ACE) Research Group, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
36
|
Llorens-Martín M. Exercising New Neurons to Vanquish Alzheimer Disease. Brain Plast 2018; 4:111-126. [PMID: 30564550 PMCID: PMC6296267 DOI: 10.3233/bpl-180065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer disease (AD) is the most common type of dementia in individuals over 65 years of age. The neuropathological hallmarks of the condition are Tau neurofibrillary tangles and Amyloid-β senile plaques. Moreover, certain susceptible regions of the brain experience a generalized lack of neural plasticity and marked synaptic alterations during the progression of this as yet incurable disease. One of these regions, the hippocampus, is characterized by the continuous addition of new neurons throughout life. This phenomenon, named adult hippocampal neurogenesis (AHN), provides a potentially endless source of new synaptic elements that increase the complexity and plasticity of the hippocampal circuitry. Numerous lines of evidence show that physical activity and environmental enrichment (EE) are among the most potent positive regulators of AHN. Given that neural plasticity is markedly decreased in many neurodegenerative diseases, the therapeutic potential of making certain lifestyle changes, such as increasing physical activity, is being recognised in several non-pharmacologic strategies seeking to slow down or prevent the progression of these diseases. This review article summarizes current evidence supporting the putative therapeutic potential of EE and physical exercise to increase AHN and hippocampal plasticity both under physiological and pathological circumstances, with a special emphasis on neurodegenerative diseases and AD.
Collapse
Affiliation(s)
- María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa”, CBMSO, CSIC-UAM, Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases CIBERNED, Madrid, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
37
|
Lin TW, Tsai SF, Kuo YM. Physical Exercise Enhances Neuroplasticity and Delays Alzheimer's Disease. Brain Plast 2018; 4:95-110. [PMID: 30564549 PMCID: PMC6296269 DOI: 10.3233/bpl-180073] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence indicates that exercise can improve learning and memory as well as attenuate neurodegeneration, including Alzheimer's disease (AD). In addition to improving neuroplasticity by altering the synaptic structure and function in various brain regions, exercise also modulates systems like angiogenesis and glial activation that are known to support neuroplasticity. Moreover, exercise helps to maintain a cerebral microenvironment that facilitates synaptic plasticity by enhancing the clearance of Aβ, one of the main culprits of AD pathogenesis. The purpose of this review is to highlight the positive impacts of exercise on promoting neuroplasticity. Possible mechanisms involved in exercise-modulated neuroplasticity are also discussed. Undoubtedly, more studies are needed to design an optimal personalized exercise protocol for enhancing brain function.
Collapse
Affiliation(s)
- Tzu-Wei Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, Georgia, USA
| | - Sheng-Feng Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
38
|
Carlson SW, Saatman KE. Central Infusion of Insulin-Like Growth Factor-1 Increases Hippocampal Neurogenesis and Improves Neurobehavioral Function after Traumatic Brain Injury. J Neurotrauma 2018; 35:1467-1480. [PMID: 29455576 PMCID: PMC5998830 DOI: 10.1089/neu.2017.5374] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) produces neuronal dysfunction and cellular loss that can culminate in lasting impairments in cognitive and motor abilities. Therapeutic agents that promote repair and replenish neurons post-TBI hold promise in improving recovery of function. Insulin-like growth factor-1 (IGF-1) is a neurotrophic factor capable of mediating neuroprotective and neuroplasticity mechanisms. Targeted overexpression of IGF-1 enhances the generation of hippocampal newborn neurons in brain-injured mice; however, the translational neurogenic potential of exogenously administered IGF-1 post-TBI remains unknown. In a mouse model of controlled cortical impact, continuous intracerebroventricular infusion of recombinant human IGF-1 (hIGF) for 7 days, beginning 15 min post-injury, resulted in a dose-dependent increase in the number of immature neurons in the hippocampus. Infusion of 10 μg/day of IGF-1 produced detectable levels of hIGF-1 in the cortex and hippocampus and a concomitant increase in protein kinase B activation in the hippocampus. Both motor function and cognition were improved over 7 days post-injury in IGF-1-treated cohorts. Vehicle-treated brain-injured mice showed reduced hippocampal immature neuron density relative to sham controls at 7 days post-injury. In contrast, the density of hippocampal immature neurons in brain-injured mice receiving acute onset IGF-1 infusion was significantly higher than in injured mice receiving vehicle and equivalent to that in sham-injured control mice. Importantly, the neurogenic effect of IGF-1 was maintained with as much as a 6-h delay in the initiation of infusion. These data suggest that central infusion of IGF-1 enhances the generation of immature neurons in the hippocampus, with a therapeutic window of at least 6 h post-injury, and promotes neurobehavioral recovery post-TBI.
Collapse
Affiliation(s)
- Shaun W. Carlson
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
39
|
Karlsson L, González-Alvarado MN, Larrosa-Flor M, Osman A, Börjesson M, Blomgren K, Kuhn HG. Constitutive PGC-1α Overexpression in Skeletal Muscle Does Not Improve Morphological Outcome in Mouse Models of Brain Irradiation or Cortical Stroke. Neuroscience 2018; 384:314-328. [PMID: 29859976 DOI: 10.1016/j.neuroscience.2018.05.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/19/2018] [Accepted: 05/23/2018] [Indexed: 12/22/2022]
Abstract
Physical exercise can improve morphological outcomes after ischemic stroke and ameliorate irradiation-induced reduction of hippocampal neurogenesis in rodents, but the mechanisms underlying these effects remain largely unknown. The transcription factor peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) is considered to be one of the central factors responsible for exercise-induced benefits in skeletal muscle, including the release of neurotrophic factors into the circulation. In order to test if PGC-1α overexpression in skeletal muscle could simulate the exercise-induced effects on recovery after cranial irradiation and stroke, we used male adult transgenic mice overexpressing murine PGC-1α under the control of muscle creatinine kinase promoter and subjected them to either whole brain irradiation at a dose of 4 Gy or photothrombotic stroke to the sensory motor cortex. Muscular PGC-1α overexpression did not ameliorate irradiation-induced reduction of newborn BrdU-labeled cells in the dentate gyrus, immature neurons, or newborn mature neurons. In the stroke model, muscular overexpression of PGC-1α resulted in an increased infarct size without any changes in microglia activation or reactive astrocytosis. No difference could be detected in the number of migrating neural progenitor cells from the subventricular zone to the lesioned neocortex or in vascular density of the contralateral neocortex in comparison to wildtype animals. We conclude that forced muscular overexpression of PGC-1α does not have a beneficial effect on hippocampal neurogenesis after irradiation, but rather a detrimental effect on the infarct volume after stroke in mice. This suggests that artificial muscle activation through the PGC-1α pathway is not sufficient to mimic exercise-induced recovery after cranial irradiation and stroke.
Collapse
Affiliation(s)
- Lars Karlsson
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden; The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, 416 85 Gothenburg, Sweden.
| | | | - Mar Larrosa-Flor
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden
| | - Ahmed Osman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Mats Börjesson
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden; Center for Health and Performance, Department of Food and Nutrition, University of Gothenburg, Box 300, 405 30 Gothenburg, Sweden; Sahlgrenska University Hospital/Östra, 416 50 Gothenburg, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Hans Georg Kuhn
- Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30 Gothenburg, Sweden
| |
Collapse
|
40
|
Selective alteration of adult hippocampal neurogenesis and impaired spatial pattern separation performance in the RSK2-deficient mouse model of Coffin-Lowry syndrome. Neurobiol Dis 2018; 115:69-81. [PMID: 29627578 DOI: 10.1016/j.nbd.2018.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/07/2018] [Accepted: 04/04/2018] [Indexed: 01/12/2023] Open
Abstract
Adult neurogenesis is involved in certain hippocampus-dependent cognitive functions and is linked to psychiatric diseases including intellectual disabilities. The Coffin-Lowry syndrome (CLS) is a developmental disorder caused by mutations in the Rsk2 gene and characterized by intellectual disabilities associated with growth retardation. How RSK2-deficiency leads to cognitive dysfunctions in CLS is however poorly understood. Here, using Rsk2 Knock-Out mice, we characterized the impact of RSK2 deficiency on adult hippocampal neurogenesis in vivo. We report that the absence of RSK2 does not affect basal proliferation, differentiation and survival of dentate gyrus adult-born neurons but alters the maturation progression of young immature newborn neurons. Moreover, when RSK2-deficient mice were submitted to spatial learning, in contrast to wild-type mice, proliferation of adult generated neurons was decreased and no pro-survival effect of learning was observed. Thus, learning failed to recruit a selective population of young newborn neurons in association with deficient long-term memory recall. Given the proposed role of the dentate gyrus and of adult-generated newborn neurons in hippocampal-dependent pattern separation function, we explored this function in a delayed non-matching to place task and in an object-place pattern separation task and report severe deficits in spatial pattern separation in Rsk2-KO mice. Together, this study reveals a previously unknown role for RSK2 in the early stages of maturation and learning-dependent involvement of adult-born dentate gyrus neurons. These alterations associated with a deficit in the ability of RSK2-deficient mice to finely discriminate relatively similar spatial configurations, may contribute to cognitive dysfunction in CLS.
Collapse
|
41
|
Morgan AH, Andrews ZB, Davies JS. Less is more: Caloric regulation of neurogenesis and adult brain function. J Neuroendocrinol 2017; 29. [PMID: 28771924 DOI: 10.1111/jne.12512] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/20/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022]
Abstract
Calorie intake is essential for regulating normal physiological processes and is fundamental to maintaining life. Indeed, both extremes of calorie intake result in increased morbidity and mortality. In this review, we discuss the effect of calorie intake on adult brain function, with an emphasis on the beneficial effects of mild calorie restriction. Recent findings relating to the regenerative and protective effects of the gastrointestinal hormone, ghrelin, suggest that it may underlie the beneficial effects of calorie restriction. We discuss the putative cellular mechanisms underlying the action of ghrelin and their possible role in supporting healthy brain ageing.
Collapse
Affiliation(s)
- A H Morgan
- Molecular Neurobiology, Institute of Life Science, School of Medicine, Swansea University, Swansea, UK
| | - Z B Andrews
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - J S Davies
- Molecular Neurobiology, Institute of Life Science, School of Medicine, Swansea University, Swansea, UK
| |
Collapse
|
42
|
Torres-Pérez M, Rosillo JC, Berrosteguieta I, Olivera-Bravo S, Casanova G, García-Verdugo JM, Fernández AS. Stem cells distribution, cellular proliferation and migration in the adult Austrolebias charrua brain. Brain Res 2017; 1673:11-22. [DOI: 10.1016/j.brainres.2017.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 12/18/2022]
|
43
|
Macpherson H, Teo WP, Schneider LA, Smith AE. A Life-Long Approach to Physical Activity for Brain Health. Front Aging Neurosci 2017; 9:147. [PMID: 28588474 PMCID: PMC5440589 DOI: 10.3389/fnagi.2017.00147] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
It is well established that engaging in lifelong Physical activity (PA) can help delay the onset of many chronic lifestyle related and non-communicable diseases such as cardiovascular disease, type two diabetes, cancer and chronic respiratory diseases. Additionally, growing evidence also documents the importance of PA for brain health, with numerous studies indicating regular engagement in physical activities may be protective against cognitive decline and dementia in late life. Indeed, the link between PA and brain health may be different at each stage of life from childhood, mid-life and late life. Building on this emerging body of multidisciplinary research, this review aims to summarize the current body of evidence linking regular PA and brain health across the lifespan. Specifically, we will focus on the relationship between PA and brain health at three distinct stages of life: childhood and adolescence, mid-life, late life in cognitively healthy adults and later life in adults living with age-related neurodegenerative disorders such as Parkinson's disease (PD) and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Helen Macpherson
- Institute for Physical Activity and Nutrition, Deakin UniversityBurwood, VIC, Australia
| | - Wei-P Teo
- Institute for Physical Activity and Nutrition, Deakin UniversityBurwood, VIC, Australia
| | - Luke A Schneider
- Robinson Research Institute, University of AdelaideAdelaide, SA, Australia
| | - Ashleigh E Smith
- Alliance for Research in Exercise Nutrition and Activity (ARENA), School of Health Sciences, University of South AustraliaAdelaide, SA, Australia
| |
Collapse
|
44
|
Argente-Arizón P, Guerra-Cantera S, Garcia-Segura LM, Argente J, Chowen JA. Glial cells and energy balance. J Mol Endocrinol 2017; 58:R59-R71. [PMID: 27864453 DOI: 10.1530/jme-16-0182] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/18/2016] [Indexed: 12/31/2022]
Abstract
The search for new strategies and drugs to abate the current obesity epidemic has led to the intensification of research aimed at understanding the neuroendocrine control of appetite and energy expenditure. This intensified investigation of metabolic control has also included the study of how glial cells participate in this process. Glia, the most abundant cell type in the central nervous system, perform a wide spectrum of functions and are vital for the correct functioning of neurons and neuronal circuits. Current evidence indicates that hypothalamic glia, in particular astrocytes, tanycytes and microglia, are involved in both physiological and pathophysiological mechanisms of appetite and metabolic control, at least in part by regulating the signals reaching metabolic neuronal circuits. Glia transport nutrients, hormones and neurotransmitters; they secrete growth factors, hormones, cytokines and gliotransmitters and are a source of neuroprogenitor cells. These functions are regulated, as glia also respond to numerous hormones and nutrients, with the lack of specific hormonal signaling in hypothalamic astrocytes disrupting metabolic homeostasis. Here, we review some of the more recent advances in the role of glial cells in metabolic control, with a special emphasis on the differences between glial cell responses in males and females.
Collapse
Affiliation(s)
- Pilar Argente-Arizón
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Santiago Guerra-Cantera
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jesús Argente
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A Chowen
- Departments of Pediatrics & Pediatric EndocrinologyHospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
45
|
Joro R, Uusitalo A, DeRuisseau KC, Atalay M. Changes in cytokines, leptin, and IGF-1 levels in overtrained athletes during a prolonged recovery phase: A case-control study. J Sports Sci 2016; 35:2342-2349. [PMID: 27966392 DOI: 10.1080/02640414.2016.1266379] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We investigated how cytokines are implicated with overtraining syndrome (OTS) in athletes during a prolonged period of recovery. Plasma IL-6, IL-10, TNF-α, IL-1β, adipokine leptin, and insulin like growth factor-1 (IGF-1) concentrations were measured in overtrained (OA: 5 men, 2 women) and healthy control athletes (CA: 5 men, 5 women) before and after exercise to volitional exhaustion. Measurements were conducted at baseline and after 6 and 12 months. Inflammatory cytokines did not differ between groups at rest. However, resting leptin concentration was lower in OA than CA at every measurement (P < 0.050) but was not affected by acute exercise. Although IL-6 and TNF-α concentrations increased with exercise in both groups (P < 0.050), pro-inflammatory IL-1β concentration increased only in OA (P < 0.050) and anti-inflammatory IL-10 was greater in CA (P < 0.001). In OA, exercise-related IL-6 and TNF-α induction was enhanced during the follow-up (P < 0.050). IGF-1 decreased with exercise in OA (P < 0.050); however, no differences in resting IGF-1 were observed. In conclusion, low leptin level at rest and a pro-inflammatory cytokine response to acute exercise may reflect a chronic maladaptation state in overtrained athletes. In contrast, the accentuation of IL-6 and TNF-α responses to acute exercise seemed to associate with the progression of recovery from overtraining.
Collapse
Affiliation(s)
- Raimo Joro
- a Institute of Biomedicine, Physiology , University of Eastern Finland , Kuopio , Finland
| | - Arja Uusitalo
- b HUS Medical Imaging Center, Clinical Physiology and Nuclear Medicine , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Keith C DeRuisseau
- a Institute of Biomedicine, Physiology , University of Eastern Finland , Kuopio , Finland.,c Department of Exercise Science , Syracuse University , Syracuse , NY , USA
| | - Mustafa Atalay
- a Institute of Biomedicine, Physiology , University of Eastern Finland , Kuopio , Finland
| |
Collapse
|
46
|
Mysoet J, Canu MH, Gillet C, Fourneau J, Garnier C, Bastide B, Dupont E. Reorganization of motor cortex and impairment of motor performance induced by hindlimb unloading are partially reversed by cortical IGF-1 administration. Behav Brain Res 2016; 317:434-443. [PMID: 27717815 DOI: 10.1016/j.bbr.2016.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/29/2016] [Accepted: 10/03/2016] [Indexed: 10/20/2022]
Abstract
Immobilization, bed rest, or sedentary lifestyle, are known to induce a profound impairment in sensorimotor performance. These alterations are due to a combination of peripheral and central factors. Previous data conducted on a rat model of disuse (hindlimb unloading, HU) have shown a profound reorganization of motor cortex and an impairment of motor performance. Recently, our interest was turned towards the role of insulin-like growth factor 1 (IGF-1) in cerebral plasticity since this growth factor is considered as the mediator of beneficial effects of exercise on the central nervous system, and its cortical level is decreased after a 14-day period of HU. In the present study, we attempted to determine whether a chronic subdural administration of IGF-1 in HU rats could prevent deleterious effects of HU on the motor cortex and on motor activity. We demonstrated that HU induces a shrinkage of hindlimb cortical representation and an increase in current threshold to elicit a movement. Administration of IGF-1 in HU rats partially reversed these changes. The functional evaluation revealed that IGF-1 prevents the decrease in spontaneous activity found in HU rats and the changes in hip kinematics during overground locomotion, but had no effect of challenged locomotion (ladder rung walking test). Taken together, these data clearly indicate the implication of IGF-1 in cortical plastic mechanisms and in behavioral alteration induced by a decreased in sensorimotor activity.
Collapse
Affiliation(s)
- Julien Mysoet
- Univ. Lille, EA 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Eurasport, 413 Rue Eugène Avinée, F-59120 Loos, France.
| | - Marie-Hélène Canu
- Univ. Lille, EA 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Eurasport, 413 Rue Eugène Avinée, F-59120 Loos, France.
| | - Christophe Gillet
- Univ. Valenciennes, LAMIH UMR CNRS 8201 - Laboratory of Industrial and Human Automation control, Mechanical engineering and Computer Science, Le Mont Houy, F-59313 Valenciennes cedex 9, France.
| | - Julie Fourneau
- Univ. Lille, EA 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Eurasport, 413 Rue Eugène Avinée, F-59120 Loos, France.
| | - Cyril Garnier
- Univ. Valenciennes, LAMIH UMR CNRS 8201 - Laboratory of Industrial and Human Automation control, Mechanical engineering and Computer Science, Le Mont Houy, F-59313 Valenciennes cedex 9, France.
| | - Bruno Bastide
- Univ. Lille, EA 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Eurasport, 413 Rue Eugène Avinée, F-59120 Loos, France.
| | - Erwan Dupont
- Univ. Lille, EA 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Eurasport, 413 Rue Eugène Avinée, F-59120 Loos, France.
| |
Collapse
|
47
|
Marwarha G, Claycombe K, Schommer J, Collins D, Ghribi O. Palmitate-induced Endoplasmic Reticulum stress and subsequent C/EBPα Homologous Protein activation attenuates leptin and Insulin-like growth factor 1 expression in the brain. Cell Signal 2016; 28:1789-805. [PMID: 27555288 DOI: 10.1016/j.cellsig.2016.08.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/05/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022]
Abstract
The peptide hormones Insulin-like growth factor-1 (IGF1) and leptin mediate a myriad of biological effects - both in the peripheral and central nervous systems. The transcription of these two hormones is regulated by the transcription factor C/EBPα, which in turn is negatively regulated by the transcription factor C/EBP Homologous Protein (CHOP), a specific marker of endoplasmic reticulum (ER) stress. In the peripheral system, disturbances in leptin and IGF-1 levels are implicated in a variety of metabolic diseases including obesity, diabetes, atherosclerosis and cardiovascular diseases. Current research suggests a positive correlation between consumption of diets rich in saturated free fatty acids (sFFA) and metabolic diseases. Induction of ER stress and subsequent dysregulation in the expression levels of leptin and IGF-1 have been shown to mediate sFFA-induced metabolic diseases in the peripheral system. Palmitic acid (palmitate), the most commonly consumed sFFA, has been shown to be up-taken by the brain, where it may promote neurodegeneration. However, the extent to which palmitate induces ER stress in the brain and attenuates leptin and IGF1 expression has not been determined. We fed C57BL/6J mice a palmitate-enriched diet and determined effects on the expression levels of leptin and IGF1 in the hippocampus and cortex. We further determined the extent to which ER stress and subsequent CHOP activation mediate the palmitate effects on the transcription of leptin and IGF1. We demonstrate that palmitate induces ER stress and decreases leptin and IGF1 expression by inducing the expression of CHOP. The molecular chaperone 4-phenylbutyric acid (4-PBA), an inhibitor of ER stress, precludes the palmitate-evoked down-regulation of leptin and IGF1 expression. Furthermore, the activation of CHOP in response to ER stress is pivotal in the attenuation of leptin and IGF1 expression as knocking-down CHOP in mice or in SH-SY5Y and Neuro-2a (N2a) cells rescues the palmitate-induced mitigation in leptin and IGF1 expression. Our study implicates for the first time ER stress-induced CHOP activation in the brain as a mechanistic link in the palmitate-induced negative regulation of leptin and IGF1, two neurotrophic cytokines that play an indispensable role in the mammalian brain.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Kate Claycombe
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Jared Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - David Collins
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA.
| |
Collapse
|
48
|
Kandola A, Hendrikse J, Lucassen PJ, Yücel M. Aerobic Exercise as a Tool to Improve Hippocampal Plasticity and Function in Humans: Practical Implications for Mental Health Treatment. Front Hum Neurosci 2016; 10:373. [PMID: 27524962 PMCID: PMC4965462 DOI: 10.3389/fnhum.2016.00373] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/11/2016] [Indexed: 12/24/2022] Open
Abstract
Aerobic exercise (AE) has been widely praised for its potential benefits to cognition and overall brain and mental health. In particular, AE has a potent impact on promoting the function of the hippocampus and stimulating neuroplasticity. As the evidence-base rapidly builds, and given most of the supporting work can be readily translated from animal models to humans, the potential for AE to be applied as a therapeutic or adjunctive intervention for a range of human conditions appears ever more promising. Notably, many psychiatric and neurological disorders have been associated with hippocampal dysfunction, which may underlie the expression of certain symptoms common to these disorders, including (aspects of) cognitive dysfunction. Augmenting existing treatment approaches using AE based interventions may promote hippocampal function and alleviate cognitive deficits in various psychiatric disorders that currently remain untreated. Incorporating non-pharmacological interventions into clinical treatment may also have a number of other benefits to patient well being, such as limiting the risk of adverse side effects. This review incorporates both animal and human literature to comprehensively detail how AE is associated with cognitive enhancements and stimulates a cascade of neuroplastic mechanisms that support improvements in hippocampal functioning. Using the examples of schizophrenia and major depressive disorder, the utility and implementation of an AE intervention to the clinical domain will be proposed, aimed to reduce cognitive deficits in these, and related disorders.
Collapse
Affiliation(s)
- Aaron Kandola
- Brain and Mental Health Lab, School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, MelbourneVIC, Australia; Amsterdam Brain and Cognition, University of AmsterdamAmsterdam, Netherlands
| | - Joshua Hendrikse
- Brain and Mental Health Lab, School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne VIC, Australia
| | - Paul J Lucassen
- Centre for Neuroscience, Swammerdam Institute of Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Murat Yücel
- Brain and Mental Health Lab, School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne VIC, Australia
| |
Collapse
|
49
|
Mooney SJ, Shah K, Yeung S, Burgess A, Aubert I, Hynynen K. Focused Ultrasound-Induced Neurogenesis Requires an Increase in Blood-Brain Barrier Permeability. PLoS One 2016; 11:e0159892. [PMID: 27459643 PMCID: PMC4961388 DOI: 10.1371/journal.pone.0159892] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/08/2016] [Indexed: 11/23/2022] Open
Abstract
Transcranial focused ultrasound technology used to transiently open the blood-brain barrier, is capable of stimulating hippocampal neurogenesis; however, it is not yet known what aspects of the treatment are necessary for enhanced neurogenesis to occur. The present study set out to determine whether the opening of blood-brain barrier, the specific pressure amplitudes of focused ultrasound, and/or the intravenous administration of microbubbles (phospholipid microspheres) are necessary for the enhancement of neurogenesis. Specifically, mice were exposed to burst (10ms, 1Hz burst repetition frequency) focused ultrasound at the frequency of 1.68MHz and with 0.39, 0.78, 1.56 and 3.0MPa pressure amplitudes. These treatments were also conducted with or without microbubbles, at 0.39 + 0.78MPa or 1.56 + 3.0MPa, respectively. Only focused ultrasound at the ~0.78 MPa pressure amplitude with microbubbles promoted hippocampal neurogenesis and was associated with an increase in blood-brain barrier permeability. These results suggest that focused ultrasound -mediated neurogenesis is dependent upon the opening of the blood-brain barrier.
Collapse
Affiliation(s)
- Skyler J. Mooney
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- * E-mail:
| | - Kairavi Shah
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Sharon Yeung
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Isabelle Aubert
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Nicastro TM, Greenwood BN. Central monoaminergic systems are a site of convergence of signals conveying the experience of exercise to brain circuits involved in cognition and emotional behavior. Curr Zool 2016; 62:293-306. [PMID: 29491917 PMCID: PMC5804240 DOI: 10.1093/cz/zow027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 02/11/2016] [Indexed: 01/04/2023] Open
Abstract
Physical activity can enhance cognitive function and increase resistance against deleterious effects of stress on mental health. Enhanced cognitive function and stress resistance produced by exercise are conserved among vertebrates, suggesting that ubiquitous mechanisms may underlie beneficial effects of exercise. In the current review, we summarize the beneficial effects of exercise on cognitive function and stress resistance and discuss central and peripheral signaling factors that may be critical for conferring the effects of physical activity to brain circuits involved in cognitive function and stress. Additionally, it is suggested that norepinephrine and serotonin, highly conserved monoamines that are sensitive to exercise and able to modulate behavior in multiple species, could represent a convergence between peripheral and central exercise signals that mediate the beneficial effects of exercise. Finally, we offer the novel hypothesis that thermoregulation during exercise could contribute to the emotional effects of exercise by activating a subset of temperature-sensitive serotonergic neurons in the dorsal raphe nucleus that convey anxiolytic and stress-protective signals to forebrain regions. Throughout the review, we discuss limitations to current approaches and offer strategies for future research in exercise neuroscience.
Collapse
|